Curr. Issues Mol. Biol. 14: 9-26. Ribonucleotide Reductase as a Target to ControlOnline Apicomplexan journal at http://www.cimb.org Diseases 9 Ribonucleotide Reductase as a Target to Control Apicomplexan Diseases

James B. Munro and Joana C. Silva* hosts/vectors and transition between life cycle stages is dependent upon a diverse array of environmental cues. Department of Microbiology and Immunology and Institute The biological characteristics that differentiate for Genome Sciences, University of Maryland School of apicomplexans from their hosts have often been Medicine, Baltimore, MD 21201, USA considered optimal targets of new therapeutics to control these eukaryotic pathogens. Alternatively, essential and strongly conserved proteins can be targeted, provided that Abstract they differ from their vertebrate homologs in such a way that Malaria is caused by species in the apicomplexan genus minimizes potential cross-reaction and toxicity. , which infect hundreds of millions of people The enzyme ribonucleotide reductase (RNR) is one each year and kill close to one million. While malaria is such example. RNR utilizes free radical chemistry to catalyze the most notorious of the apicomplexan-caused diseases, the reduction of ribonucleotides to deoxyribonucleotides other members of the eukaryotic phylum (Thelander and Reichard, 1979; Reichard, 1988). It provides are responsible for additional, albeit less well-known, the only de novo means of generating the essential building diseases in humans, economically important livestock, and blocks for DNA replication and repair across all domains a variety of other . Diseases such as babesiosis of life and, as such, it is the rate-limiting step in DNA (hemolytic anemia), theileriosis and East Coast Fever, synthesis (Jordan and Reichard, 1998; Lundin et al., 2009). , and toxoplasmosis are caused by the Additionally, RNR is critical for maintaining a balanced pool apicomplexans , , and of DNA precursors during chromosome replication (Herrick Toxoplasma, respectively. In addition to the loss of human and Sclavi, 2007). An unbalanced deoxyribonucleotide life, these diseases are responsible for losses of billions triphosphate pool may lead to an increase in mutation and of dollars annually. Hence, the research into new drug disease (Lin and Elford, 1980; Reichard, 1988; Chabes et targets remains a high priority. Ribonucleotide reductase al., 2003; Wheeler et al., 2005; Gon et al., 2006; Mathews, (RNR) is an essential enzyme found in all domains of 2006; Kumar et al., 2010). life. It is the only means by which de novo synthesis of Here we review the chemotherapeutic methods used to deoxyribonucleotides occurs, without which DNA replication inhibit the essential enzyme RNR, with particular emphasis and repair cannot proceed. RNR has long been the target of on the novel RNR R2_e2 subunit recently identifed in antiviral, antibacterial and anti-cancer therapeutics. Herein, apicomplexan parasites (Munro et al., submitted) and on the we review the chemotherapeutic methods used to inhibit malaria-causing genus Plasmodium. The R2_e2 subunit is RNR, with particular emphasis on the role of RNR inhibition unique to the Apicomplexa and as such, it can potentially be in Apicomplexa, and in light of the novel RNR R2_e2 subunit used to specifcally target apicomplexan pathogens. recently identifed in apicomplexan parasites. Apicomplexan parasites are responsible for devastating Introduction infectious diseases The Apicomplexa are a group of single-celled, eukaryotic The phylum Apicomplexa consists of more than 4,000 organisms that, together with the and dinofagelates, described species (Levine, 1988), many of which are of form the major lineages in the . All Apicomplexa, medical, agricultural, and economic importance and whose save the predatory fagellates , are pathogenetic, adverse impact on human society cannot be overstated. obligate, intracellular parasites (Adl et al., 2005; Morrison, Among the most notorious are Plasmodium, Babesia, 2009). They are characterized by the presence of an apical Theileria, Cryptosporidium, and Toxoplasma the causative complex, a structure involved in host-cell invasion, which is agents of malaria, babesiosis, theileriosis and East Coast located at the anterior end of the cell. Most apicomplexans fever, cryptosporidiosis, and toxoplasmosis, respectively. also possess a specialized organelle called the apicoplast, They are responsible for causing millions of human deaths a secondary endosymbiotic plastid believed to be of red and billions of dollars in productivity and material losses algal origin (Blanchard and Hicks, 1999; Fast et al., 2001; each year (Sachs and Malaney, 2002; Corso et al., 2003; Janouškovec et al., 2010). Within the apicoplast occur Rowe et al., 2006; Spielman, 2009). Currently, fve species processes essential for the parasite's survival, such as of Plasmodium are known to cause malaria in humans, P. heme and lipid biosynthesis. Another defning characteristic falciparum, P. knowlesi, P. malariae, P. ovale, and P. vivax of apicomplexans is their inability to synthesize purine rings (Rougemont et al., 2004; Singh et al., 2004; Cox-Singh et de novo and hence their need to salvage exogenous purines al., 2008), of which P. falciparum is the most deadly and via a variety of different pathways (Booden and Hull, 1973; P. vivax the most geographically widespread. The life cycle Chaudhary et al., 2004; de Koning et al., 2005; Cassera of Plasmodium alternates between a vertebrate host and et al., 2008; Madrid et al., 2008). These organisms have mosquito vector and involves four major developmental complex (indirect) life cycles, and they often exploit multiple stages in the vertebrate host: sporozoites, merozoites, trophozoites, and gametocytes (Bledsoe, 2005; Brown and Catteruccia, 2006). *Corresponding author: [email protected]

Horizon Scientifc Press. http://www.horizonpress.com 10 Munro and Silva

Prioritization of apicomplexan drug targets been proposed as a means to combat the emergence of There is currently no fully effcacious vaccine on the market drug resistance (Durand et al., 2008). Genes with high rates against any apicomplexan species and drug treatment is of nonsynonymous changes have been associated with the method of choice in the management of apicomplexan drug resistance in P. vivax (Dharia et al., 2010), and may be diseases. Modern approaches to drug design, made responsible for vaccine evasion in P. falciparum (Takala and possible with the advent of genome sequencing, emphasize Plowe, 2009). In contrast, evolutionary patterning focuses defning and targeting metabolic and molecular differences on fnding and targeting protein residues that are under between host and parasite to avoid host side effects (Croft, strong purifying selection, which will in principle reduce the 1997). This may be achieved with the selective targeting of instances of drug resistance mutations. parasite-specifc enzymes or by targeting those which are The tremendous health burden imposed by malaria highly divergent or have distinct binding sites and are thus has made Plasmodium the primary target for many of these suffciently different to be selectively targeted (Coombs, approaches. Despite a diversifed arsenal of potential tools 1999; Cerqueira et al., 2007). Also important is that the to combat malarial infection, multiple drug resistance to protein target be essential to the growth, reproduction, or existing anti-malarial compounds is becoming increasingly survival of the parasite. Finally, knowledge of the gene common in Plasmodium (Greenwood and Mutabingwa, expression pattern of potential target proteins is necessary to 2002; Anderson, 2009; Bustamante et al., 2009; Takala and link drug administration with the critical and relevant stages Plowe, 2009). A case in point is the antifolate drugs used to of the pathogen's life cycle. This is particularly pertinent to treat malaria. Antifolate drugs bind enzymes necessary for organisms characterized by differentially expressed, stage- folate biosynthesis, thus targeting essential precursors for specifc, and often stage-unique gene expression, such as purine and pyrimidine synthesis. Antifolates have been used apicomplexans (Coulson et al., 2004). It has been suggested against Plasmodium with success, but resistance to these that targeting the intraerythrocytic life stages of Plasmodium drugs has become widespread (Gregson and Plowe, 2005; (the ring forms, trophozoites, schizonts, and merozoites), Mkulama et al., 2008; Sridaran et al., 2010). Currently, the when clinical symptoms are manifested, is of particular primary treatment for malaria is based on artemisinin, which interest (Yeh and Altman, 2006). However, targeting multiple is administered in combination with other drugs in order to life stages, for example controlling both the liver and blood prevent, or delay, the onset of resistance. However, there stages of Plasmodium, may be more conducive to the are clear indications that artemisinin resistance is emerging ultimate goal of disease eradication (Alonso et al., 2011). (Chrubasik and Jacobson, 2010; Dondorp et al., 2010; Enserink, 2010; Fidock, 2010). Therefore, the development Current control strategies and challenges of new chemotherapeutic and prophylactic antimalarial Drug targets for control of apicomplexans have focused on drugs and vaccines remains a priority (Greenwood and parasite metabolic processes. These include processes Mutabingwa, 2002; Anderson, 2009; Bustamante et al., within the cytosol, mitochondrion, digestive vacuole, 2009; Takala and Plowe, 2009). synthesis of macromolecular and metabolic enzymes, and processes involved in membrane synthesis and signaling RNR classifcation and distribution (Olliaro and Yuthavong, 1999; Padmanaban, 2003; Fidock RNRs are classifed into one of three classes, I-III (Jordan et al., 2004; El Bissati et al., 2006). Enzymes in the purine et al., 1994; Reichard, 1997; Fontecave, 1998; Nordlund salvage pathways, essential to these pathogens, are also and Reichard, 2006). All three classes use a thiyl radical potential drug targets (Tracy and Sherman, 1972; Krug to remove the ribose OH-group. The distinction between et al., 1989; Parker et al., 2000; Gardner et al., 2002; these classes relies on differences in radical generation Raman and Balaram, 2004; Striepen et al., 2004; Ting chemistry and the cofactor needed to produce the organic et al., 2005; Downie et al., 2008), as are all proteins and radical. Class I RNRs are oxygen-dependent, typically processes related to the apicomplexan-specifc organelle, require a tyrosyl radical and a diiron center, and are the apicoplast (Foth et al., 2003; Sato and Wilson, 2005; characteristic of and common among bacteria. Wiesner et al., 2008; Lizundia et al., 2009). Antibiotics like Class II RNRs are indifferent to oxygen, form a thiyl radical doxycycline, which specifcally target plastid pathways and via adenosylcobalamin, and are characteristic of Archaea impair the expression of apicoplast genes, can be used and bacteria. Class III RNRs are anaerobic, form a glycyl to target apicomplexans (Ralph et al., 2001; Leander and radical using an iron-sulfur center in the presence of Keeling, 2003; Dahl et al., 2006). Apicoplast drug targets S-adenosylmethionine and reduced favodoxin, and are have included the apicoplast's metabolic pathways (e.g. also characteristic of Archaea and bacteria. DNA replication, transcription, protein translation, fatty acid Class I RNRs have been further categorized as class biosynthesis, and isoprenoid biosynthesis), or targeting Ia, Ib, or Ic. Class Ia is typical in eukaryotes and bacteria, proteins encoded by the host's nuclear genes that are while bacteria and Archaea primarily encode classes Ib and destined for the apicoplast (McFadden and Roos, 1999; Ic RNRs (Harder, 1993; Sjöberg, 2010). Class Ic RNRs Roos et al., 1999; Roos et al., 2002; Ralph et al., 2004; are further categorized as R2c proteins and while the White, 2004; Waller and McFadden, 2005; Dahl and R2-homolog R2lox proteins are described as “R2c-like”, Rosenthal, 2008; Prusty et al., 2010). they are not believed to form active RNR holoenzymes The availability of genome sequences from several (Högbom et al., 2004; Andersson and Högbom, 2009). species and isolates of Plasmodium and other Apicomplexa This classifcation, based on structural and chemical facilitates the identifcation of novel, potential drugs for the properties, has shortcomings since classes Ia and Ic are control of apicomplexan parasites (Doolan et al., 2003; Yeh not monophyletic clades, i.e., discrete, mutually exclusive et al., 2004; Carvalho and Ménard, 2005; Winzeler, 2008; groups, which contain a most recent common ancestor and Mu et al., 2010). For example, evolutionary patterning has all of its descendants. Instead, class Ia is polyphyletic and Ic Ribonucleotide Reductase as a Target to Control Apicomplexan Diseases 11 is paraphyletic. This problem was been addressed by Munro and two allosteric effector-binding sites. dATP and ATP bind et al. (submitted) (Figure 1) who recognized four distinct to the frst allosteric site, the A-site, and serve as inhibitor class I RNR clades, namely the eukaryotic-specifc clades and stimulator, respectively, while binding of dATP, ATP, R2_e1 and R2_e2, and the clades containing primarily dGTP and dTTP to the S-site determines substrate-binding archaeal and bacterial RNRs, namely R2_ab and R2c. The preference (Brown and Reichard, 1969; Reichard et al., study also revealed that the newly discovered clade R2_e2 2000). Allosteric regulation is accomplished by changes is unique to the Apicomplexa (Munro et al., submitted). In in the conformation of loop 2 which spans both the A- and fact, the most signifcant mammalian-infecting Apicomplexa S-sites, and which is determined by the molecule bound to genera, such as Plasmodium, Cryptosporidium, and the A-site (Reichard, 2010). Reviews and additional details Babesia, all encode one R2_e2 subunit. can be found in (Eriksson et al., 1979; Reichard et al., 2000; Reichard, 2002; Crona et al., 2010; Logan, 2011). Class Ia holoenzyme regulation and formation Subunit R2, the smaller subunit, contains an amino acid Apicomplexans encode Class Ia RNRs, which are thus the residue that harbors the organic radical (Nordlund et al., focus of this review. Class Ia RNR enzymes are composed 1990; Nordlund and Eklund, 1993). A long-range electron- of two distinct subunits, R1 and R2. Subunit R1, the larger coupled pathway connects the R2 radical to a cysteine of the subunits, contains a catalytic site (substrate binding) in R1 (the thiyl radical) via hydrogen-bonded amino acid residue side chains (Nordlund et al., 1990; Stubbe et al., 2003; Kolberg et al., 2004). Binding of the R2 subunit to R2lox the R1 subunit involves the C-terminus residues of the "    R2 subunit interacting with a hydrophobic cleft in the R1 "    subunit and it has been suggested that oligomerization of "!!   R1 is a prerequisite (Climent et al., 1992; Rova et al., 1999; "! Uppsten et al., 2006). " !     The most current model for RNR in eukaryotes suggests a holoenzyme with eight subunits and is of the form α6β2, R2c where alpha stands for the R1 subunit and beta for R2 "    (Rofougaran et al., 2006). It has been proposed that in the "       absence of the effectors dATP, ATP, dGTP and dTTP, the "!!   R1 subunit is an inactive monomer; however, once dTTP "! or dGTP are bound to the S-site, an α2β2 heterodimer is formed (Ingemarson and Thelander, 1996). With increasing dATP concentration (which induces enzyme inhibition), R1 R2_ab monomers form dimmers and eventually inactive hexamers "    (formation of intermediate tetramers remains in question), "  while in the presence of the enzyme activator ATP, the "!!   holoenzyme adopts an α6β2 conformation (Fairman et al., "! 2011).

Paralogous copies of the R2 subunit R2_e1 Many genomes encode two or more distinct " !   copies of the small R2 subunit (Lundin et al., 2009). For "    example, humans have the R2 and p53R2 paralogs "!!   and S. cerevisiae the Y2 and Y4 paralogs. It is clear that "! these copies have different functional roles, be it de novo creation of deoxyribonucleotides, maintenance of the R2_e2 deoxyribonucleotide pool, mitochondrial DNA replication, " !   or DNA damage repair (Elledge and Davis, 1990; Huang "  and Elledge, 1997; Tanaka et al., 2000; Lin et al., 2004; "!!   Bourdon et al., 2007). In such instances, a ββ' confguration "! is believed to contribute to the active holoenzyme, although ββ and β'β' confgurations have been reported (Wang et al., 1997; Nguyen et al., 1999; Chabes et al., 2000; Ge et al., Figure 1. Unrooted phylogenetic relationships between the 2001; Guittet et al., 2001; Voegtli et al., 2001; Perlstein et RNR class Ia and Ic subunits and the R2lox R2 homolog al., 2005; Ortigosa et al., 2006). proteins (Munro et al., submitted). Class Ic includes the While most eukaryotes encode two R2 genes belonging R2c proteins; however, this classifcation proved to be to the typical eukaryotic clade R2_e1, apicomplexans encode paraphyletic as it failed to include the clade of proteins one R2_e1 subunit and one R2_e2 subunit. Toxoplasma now designated as R2_ab. Former class Ia proved to be appears to be an exception, as so far two R2_e1-encoding polyphyletic, including the novel R2_ab clade, which does genes have been identifed but no R2_e2 has been found in not share a recent common ancestor with the monophyletic its genome. R2_e1 and R2_e2 clade. Reference to the clades R2c, The conservation of functionally important R1 active R2_ab, R2_e1, and R2_e2 now allows for unambiguous site cysteines, and electron transfer cysteine and tyrosine reference to these RNR subunits. residues, as well as the conservation of R2 residues involved 12 Munro and Silva in iron binding, electron transport, free radical transfer, subunit had tumor-suppressing activity. Cao et al. (Cao et and the formation of the hydrophobic pocket around the al., 2003) utilized a recombinant adenovirus that encoded radical, implies that both eukaryotic pathogens and their and over-expressed the human R1 gene, which reduced hosts utilize the same free radical chemistry to synthesize proliferation of human colon adenocarcinoma cells, yet had deoxyribonucleotides (Hofer et al., 1997; Roshick et al., no effect on normal cells. On the other hand, inhibition of 2000; Akiyoshi et al., 2002; Shao et al., 2006). As such, the R2 subunit may have an antineoplastic effect, serving it would appear that targeting apicomplexan RNR by to inhibit and combat the development of cancer cells. chemotherapeutic means might have an adverse effect on Expression of R2 in conjunction with activated oncogenes the human host. This is not necessarily so. impacts a cell's malignant potential (Fan et al., 1998; Desai While prokaryotic and eukaryotic R1 and R2 subunits are et al., 2005). Overexpression of R2 is linked to increased highly conserved at, and around, the functionally important drug resistance and increased invasive potential in cancer residues (Chakrabarti et al., 1993; Sjöberg, 1997; Roshick et cells (Yen, 2003). al., 2000; Voegtli et al., 2001; Högbom et al., 2004; Högbom, RNR inhibition has been applied to the control of viruses 2010), there is considerable variation in the sequences at (Gaudreau et al., 1987; Moss et al., 1993; Bianchi et al., both the N- and C-termini (Ingram and Kinnaird, 1999). 1994; Szekeres et al., 1997; Robins, 1999), bacteria (Yang In particular, the eukaryotic orthodox R2 subunit, R2_e1, et al., 1997; Mdluli and Spigelman, 2006; Ericsson et al., presents distinct differences between apicomplexans and 2010; Lou and Zhang, 2010; Torrents and Sjöberg, 2010), mammals, including differences in key functional regions of and certain cancers (Cory, 1988; Nocentini, 1996; Gwilt the R2 protein; perhaps most notable are those differences and Tracewell, 1998). Because inhibition of RNR ceases, between C-terminal sequences (Bracchi-Ricard et al., or severely reduces, DNA replication, it has long been 2005). Furthermore, and more pertinent to this review, is considered an ideal target for the control of pathogens. As the fact that the apicomplexan-specifc R2 subunit, R2_e2, such, inhibition of RNR to control eukaryotic pathogens has offers additional unique regions for drug-targeted inhibition also been suggested (Dormeyer et al., 1997; Ekanem, 2001), (Munro et al., submitted). It is these differences between in particular those belonging to Apicomplexa (Chakrabarti et apicomplexan and mammalian host sequences that may al., 1993; Barker et al., 1996; Akiyoshi et al., 2002). In fact, best be exploited when designing chemotherapeutic drugs RNR was included in a set of 57 “gold standard” essential to specifcally target the Apicomplexa, making RNR an enzymes with experimentally documented antimalarial appealing option as a drug target. effects (Huthmacher et al., 2010). These, and other studies, have resulted in a considerable array of approaches to RNR has a long history as chemotherapeutical target inhibit RNR, which we briefy describe next. Much research has focused on the relationship between the class Ia R1 and R2 subunits in the context of human cancer. Methods of RNR inhibition It has been hypothesized that normal or over-expression of RNR may be targeted at the translational or protein levels. R1 results in suppression of malignant cells (Yen, 2003) RNR inhibitors are loosely categorized as those that prohibit and Fan et al. (Fan et al., 1997) demonstrated that the R1 the formation of an active holoenzyme or those that inhibit the

translation inhibitor prevent translation of mRNA       no subunits formed "RNAi "antisense oligonucleotides

subunits formed

dimerization inhibitor prevent holoenzyme formation     no holoenzyme formed "  sequences

holoenzyme formed catalytic & allosteric inhibitors interfere with holoenzyme "nucleoside analogues (R1) holoenzyme inactivated "allosteric inhibitors (R1) holoenzyme "     "   

Figure 2. Means of RNR inhibition. A fowchart showing the progression from mRNA to the formation of the holoenzyme (ovals) and how translation, dimerization and catalytic and allosteric inhibitors act along this process. Ribonucleotide Reductase as a Target to Control Apicomplexan Diseases 13 function of an already fully-formed holoenzyme (Cerqueira the silencing of target genes with chemically synthesized et al., 2007). At the translation level, synthesis of the enzyme small interfering RNA (siRNA) (Lee and Sinko, 2006; Vaish subunits is blocked, while at the protein level, inhibitors may et al., 2010), which typically utilizes much shorter lengths be employed to prevent the formation of the holoenzyme of double stranded RNA (20 to 25 bp). siRNA has proven or inhibitors may be used to inactivate either the R1 or R2 useful in the suppression of p53R2 expression, leading to subunit, or both (Cerqueira et al., 2005) (Figure 2). The use the inhibition of tumor growth and an increase in sensitivity of ribozymes, single strand antisense oligonucleotides, and to anticancer drugs (Yanamoto et al., 2005). small interfering RNA (siRNA) can all be defned as anti- Antisense RNA has been documented in Plasmodium mRNA strategies, or subunit synthesis inhibitors (see (Aboul- (Militello et al., 2005) and there are numerous examples Fadl, 2005) for a review of the implementation, optimization, where RNAi has reportedly been successfully used to silence and practical application of these methodologies), while genes in Plasmodium (Kumar et al., 2002; Malhotra et al., dimerization, catalytic, and allosteric inhibitors focus on the 2002; McRobert and McConkey, 2002; Mohmmed et al., inhibition of formed proteins. 2003; Dasaradhi et al., 2005; Gissot et al., 2005; Crooke et al., 2006; Sunil et al., 2008; Tuteja et al., 2008; Sriwilaijaroen Subunit synthesis inhibitors et al., 2009). However, in stark contrast to these fndings, Ribozymes are those where support for RNAi in Plasmodium is lacking. Ribozymes are catalytic RNA molecules with distinct In fact, using both an experimental and bioinformatics three-dimensional confgurations, which principally exhibit approach, Baum et al. (Baum et al., 2009) suggested that trans-cleavage properties. Ribozymes can be specifcally Plasmodium lacks RNAi functionality and the conserved designed to cleave a targeted RNA sequence, thereby enzymes necessary for RNAi activity such as Dicer and inactivating gene transcripts (Haseloff and Gerlach, 1988; Argonaute-like proteins, or their analogs. It has further been Norris et al., 2000; Citti and Rainaldi, 2005). Ribozymes and suggested that documented RNAi activity in Plasmodium their applications have been extensively reviewed (Puerta- may be the result of general toxicity to the introduced RNA, Fernández et al., 2003; Nayak and Kohli, 2005; Khan, 2006; or an alternative (non-RNAi) antisense mechanism, and Tedeschi et al., 2009). Ribozymes offer a productive avenue not the result of specifc gene targeting by RNAi (Ullu et for gene therapy and have been designed for use against al., 2004). Additional authors have also questioned RNAi inborn metabolic disorders, viral infections, and cancer activity or the presence of a classical RNAi pathway in (Lewin and Hauswirth, 2001). Apicomplexa, particularly in Plasmodium (Aravind et al., Both in vitro and in vivo studies demonstrated promising 2003; Blackman, 2003; Cerutti and Casas-Mollano, 2006; use of ribozymes to target the survivin gene, which when Xue et al., 2008). Further calling into question the utility of expressed, leads to cell proliferation, typical in most human RNAi is the fact that these studies show down-regulation, carcinomas (Choi et al., 2003). Ribozymes designed to target but not the elimination, of gene function, and the degree mouse telomerase RNA were successfully administered to which protein expression is suppressed depends of a and systemically expressed in vivo, and served to reduce variety of factors (Brown and Catteruccia, 2006). the metastatic progression of B16_F10 murine melanoma metastases (Nosrati et al., 2004). In vitro targeting of RhoC RNA antisense oligonucleotide inhibitors by ribozymes showed reduction of invasiveness in human Antisense oligonucleotides (AOs) are short (10 to 30 breast cancer cells and thus demonstrated the utility of nucleotides), single strands of RNA or DNA that serve ribozymes in gene therapy (Lane et al., 2010). Similarly, to inhibit gene expression. The sequence of an AO is ribozymes designed for targeting specifc sites for cleavage complementary to a chosen target mRNA sequence, to in human telomerase RNA were demonstrated to be effective which it will bind via canonical Watson-Crick base pairing. in arresting cell growth and induction of spontaneous cell A variety of modifcations to antisense oligonucleotides apoptosis in colon cancer cells (Lu et al., 2011). may be employed to prevent degradation, increase In the context of apicomplexan control, ribozymes affnity and potency, and to reduce non-target toxicity were successfully used to reduce malarial viability up to (Chan et al., 2006; Sahu et al., 2007; Li et al., 2010). 55% when targeting P. falciparum-specifc inserts in the Further improvements come in terms of selective delivery carbamoyl-phosphate II synthetase gene (Flores et al., systems for oligonucleotides (Ming et al., 2010). AOs may 1997). Accordingly, C-terminus insertions present in the knockdown a target mRNA molecule by means of three R1 subunit enzyme of Plasmodium and Theileria offer sites distinct processes: (1) steric inhibition, where protein uniquely different from those of their hosts, which may be translation is inhibited once AOs are bound to the target specifcally targeted by ribozymes (Ingram and Kinnaird, mRNA, (2) the non-specifc endonuclease, ribonuclease H 1999). (RNase H), may be activated and catalyze the cleavage of a DNA/mRNA duplex; alternatively, (3) pre-mRNA targeting, RNA interference which includes inhibition of splicing, inhibition of the 5' cap RNA interference (RNAi) utilizes segments of double- formation, or de-stabilization of the pre-mRNA, would inhibit stranded RNA to interfere with gene expression and it mRNA maturation (Ho et al., 1996; Baker and Monia, 1999; usually relies on the enzyme Dicer and the RNA-induced Achenbach et al., 2003; Sahu et al., 2007). silencing complex (Scherr et al., 2004). Theoretically, the Genes encoding both the large R1 RNR subunit and chemotherapeutic applicability of RNAi, and that of variants small R2 subunit have been targeted with antisense inhibition. on the RNAi theme, is extensive; however, caution is Inhibition of expression of the Herpes simplex virus was advised as there are safety and specifcity concerns (Grimm achieved using AOs to target the R1 translation initiation and Kay, 2007). There have been recent advances in the site (Aurelian and Smith, 2000). The R2-directed AO, GTI- reduction of non-target effects and improved specifcity in 2040, has shown promising selectivity and specifcity in its 14 Munro and Silva antitumor activity against a variety of human cancers (Lee et et al., 1998; Guittet et al., 1999). Hydroxyurea was shown to al., 2003; Desai et al., 2005). AOs were designed to target stop DNA synthesis in P. falciparum (Inselburg and Banyal, both the R1 and R2 subunits in oropharyngeal KB cancer 1984). Improved control of malaria utilizing a combination cells; however, only the targeted inhibition of R2 expression therapy of erythropoietin and iron sulfate in conjunction with reduced enzyme activity and inhibited cell growth (Chen et hydroxeurea has been hypothesized (Saei and Ahmadian, al., 2000). 2009). In contrast to the scavenging of radicals, iron AOs have also proven to be effective against a variety of chelators, which may destroy or prevent the formation of gene products in Plasmodium (Rapaport et al., 1992; Barker the radical (Nyholm et al., 1993; Richardson, 2002; Hodges et al., 1998; Gardiner et al., 2000; Patankar et al., 2001; et al., 2004; Whitnall et al., 2006), do not necessarily cause Kyes et al., 2002; Noonpakdee et al., 2003; Gunasekera permanent inhibition. Iron chelators target cellular iron, et al., 2004). RNR activity was inhibited by targeting the leading to iron deprivation, which has been suggested to region surrounding the translation initiation codon with AO result in RNR inhibition (Pradines et al., 1996). Iron chelators phosphorothioates for the P. falciparum R2_e1 subunit, have been demonstrated to be effective against the P. (Chakrabarti et al., 1993). falciparum trophozoite and ring stages, which, unlike host cells, demonstrated a limited to irreversible loss of capacity Protein inhibitors for recovery after the chelator was removed (Lytton et al., Dimerization (polymerization) inhibitors 1994). Dimerization inhibitors bind to one or more partners in a Substrate analogs are also referred to as suicide protein-protein interaction and hence prevent formation inhibitors and were recently reviewed (Perez et al., 2010). of the holoenzyme. In the case of RNR, they are small They are recognized as “normal” ribonucleotide substrates; peptidomimetic sequences that mimic the R2 subunit however, their interaction with the holoenzyme's active C-terminal residues. As such, they competitively bind to the site leads to inactivation of the enzyme. A case in point hydrophobic cleft in the R1 subunit, to the exclusion of R2, is inactivation of the R1 active site by use of nucleoside- and prevent formation of the holoenzyme (Paradis et al., diphosphate analogs, which bind and result in alkylation 1988; Gaudreau et al., 1990; Yang et al., 1990; Cosentino of the protein (Pereira et al., 2004; Pereira et al., 2006). et al., 1991). It is the differences between host and parasite Note that some substrate analogs such as gemcitabine and R2 C-terminal sequences that have so far lent themselves tezacitabine have additional inhibitory effects on the R2 to specifc targeting of a parasite's RNR. subunit (Salowe et al., 1993; Shao et al., 2006). RNR enzyme activity was inhibited in the Herpes simplex As noted earlier, nucleoside-triphosphates bind to the virus with the introduction of a peptide that corresponded to allosteric effector sites and serve to either activate or inhibit the C-terminus amino acid residues of the viral R2 subunit the reduction of nucleoside diphosphates (Thelander and (Gaudreau et al., 1992; Liuzzi et al., 1994). In addition to Reichard, 1979). Allosteric effector analogs, which are the Herpes simplex virus, RNR dimerization inhibitors have typically nucleoside-triphosphate analogs, thus interact with been extensively studied in E. coli, hamster, mouse, yeast, the two R1 allosteric effector-binding sites, i.e. the A- and and human cells (Cohen et al., 1986; Dutia et al., 1986; S-sites. dATP normally acts as an inhibitor; however, some Climent et al., 1991; Cosentino et al., 1991; Fisher et al., deoxyadenosine analogs have an even more powerful 1993; Davis et al., 1994). affect due to their higher affnity for the R1 effector site Likewise, in the case of apicomplexans, it is the (Cory and Mansell, 1975; Harrington and Spector, 1991; difference in the C-terminal sequences of the R2 subunits Jeha et al., 2004; Shao et al., 2006). Interference of the between parasites and their hosts that may be best exploited allosteric binding sites will have an infuence on the activity by chemotherapeutic means (Chakrabarti et al., 1993; and substrate binding affnity of the R1 subunit. While the Fisher et al., 1993; Cerqueira et al., 2005). Peptidomimetic structure of the allosteric sites in the R1 subunit may be inhibitors based on the C-terminus of the small subunit have similar between the RNR of an eukaryotic parasite and that been proposed as a means of disrupting the formation of of its host, there are usually unique substitutions between the RNR heterodimer complex in P. falciparum (Bracchi- the two, which in turn may lead to differences in allosteric Ricard et al., 2005). In fact, targeting malarial RNR activity regulation. That is the case of the RNR of humans and of P. falciparum-infected erythrocytes was accomplished by of , the causative agent of sleeping use of synthetic peptidomimetic peptides, which prevented sickness (Hofer et al., 1997). Chakrabarti et al. (Chakrabarti binding of the R1 and R2 subunits (Rubin et al., 1993). et al., 1993) suggested that differences in the N-terminus sequence of the R1 subunit of P. falciparum might indicate Catalytic and allosteric inhibitors that it too utilizes a different allosteric regulation mechanism Catalytic protein inhibitors target the active RNR holoenzyme relative to humans. The authors suggested that conservation and may act on either the R1 or R2 subunits, or both. across other protozoans, in terms of the residues whose Catalytic inhibitors may function by a variety of means, be function it is to bind dTTP, indicates that they too may it: (1) the destruction of the R2 subunit radical by radical employ an allosteric regulation mechanism that differs from scavengers or iron chelators, (2) inactivation of the R1 mammalian hosts. Such a difference has the potential to subunit active site, or (3) via substrate/nucleoside analogs, be exploited via suicide substrate inhibitors or nucleoside thus primarily acting on the R1 subunit. Allosteric inhibitors, analogues (Ingram and Kinnaird, 1999). which are also nucleoside analogs, target the R1 effector binding sites. Challenges to using RNR to control apicomplexan Radical scavengers such as hydroxyurea, irreversibly parasites destroy the tyrosol radical of the R2 protein (Krakoff et al., First and foremost, the function of the apicomplexan-specifc 1968; Lepoivre et al., 1991; Szekeres et al., 1997; Fontecave R2_e2 subunit remains unknown and, in particular, whether Ribonucleotide Reductase as a Target to Control Apicomplexan Diseases 15 this subunit is essential to the formation of a functional Human R2_e1 RNR holoenzyme has yet to be determined. Munro et al. A 1.0 (submitted) have identifed considerable variability among apicomplexans in the amino acid residue that purportedly 0.5 S harbors the free radical in the R2_e2 subunit, as well as in probability additional residues of functional importance. For example, 0.0 NVFTLDADF the lack of conservation of one of the two phenylalanine F7 F1 residues in the C-terminus (Figure 3A) raises the possibility Apicomplexan R2_e1 of a difference in the interaction between the R2_e2 and 1.0 R1 subunits relative to that observed with R2_e1. Following C T the residue notation of Fisher et al. (Fisher et al., 1993), 0.5 V AD 1 7 S N L

probability L the C-terminal residues F and F appear to be particularly D IK N S 0.0 Q TD E infuential in dictating the interaction/binding-specifcity to F F 7 1 subsites of the R1 subunit (Pellegrini et al., 2000; Pender et F F al., 2001). While the R2_e2 C-terminal residue equivalent Apicomplexan R2_e2 to F1 is maintained as phenylalanine and conserved 1.0 7 K across R2_e2, the residue equivalent to F is instead S substituted for isoleucine in all sequences sampled, save 0.5 Q TF E L

probability D V DS the cryptosporidians. This may, however, not be a concern; SFLTNEQ 7 0.0 I it has been established that F need not be stringently DF F7 F1 conserved because the R1 subsite interacting with this R2 subunit position can accommodate a variety of hydrophobic R2_e2 residues (Pender et al., 2001; Gao et al., 2002; Cooperman, B Bab_bovis_XP_001610573 SISFNEDF Bab_equi_6m007985 EIKFDEDF 2003). Also, Tyr370 in mouse R2 was determined to be The_annulata_XP_953574 DIKFDEDF essential in the radical transport pathway (Rova et al., The_parva_XP_766717 EIKFDEDF Pla_berghei_Pdb_121420 QITFDEDF 1999) and yet an equivalent to this residue is lacking in the Pla_yoelii_XP_727957 QITFDEDF apicomplexan-specifc R2_e2 subunit. Pla_chabaudi_PCAS_121490 QITFDEDF There are further challenges to RNR chemotherapy. Pla_falciparum_XP_001347439 QILFDEDF Pla_knowlesi_XP_002258799 QIVLDEDF Two widely used anti-cancer RNR inhibitors, hydroxyurea Pla_vivax_XP_001614470 QIVLDEDF and gemcitabine, are toxic to humans (Banach and Williams, Cry_hominis_XP_665685 QFSTSQDF 2000; Santini et al., 2000). Additionally, resistance to RNR Cry_muris_XP_002140092 QFSTSQDF R2_e2 only .: .:** inhibition by some radical scavengers and iron chelators has been observed (Nocentini et al., 1990; Sneeden and Loeb, R2_e1 2004; Fu and Xiao, 2006). Further concerns with regard to Hom_sapiens_isoform_2_NP_001025(p53R2) NSFTLDADF Hom_sapiens_isoform_1_NP_056528 NVFTLDADF the use of ribozymes include method of delivery, ribozyme Bab_bovis_XP_001610982 QKFATDADF stability, the secondary and tertiary structure of the target Bab_equi_6m007342 QKFSTDIEF Pla_berghei_Pdb_103660 QVFSLNTDF mRNA, and how these factors relate to accessibility of the Pla_chabaudi_XP_739266 QVFSLNTDF region being targeted for cleavage (Turner, 2000). Pla_yoelii_XP_723858 QVFSLNTDF Pla_falciparum_XP_001348226 QVFCLNTEF Pla_reichenowi_novel_model_330 QVFCLNTEF Bolstering support for use of RNR inhibition to control Pla_gallinaceum_rna_PF_0053_1_1cds QVFCLNTDF apicomplexan parasites Pla_knowlesi_XP_002260936 QVFCLNSDF Since its discovery in 1961 (Reichard et al., 1961), Pla_vivax_XP_001616894 QVFCLNSDF Cry_hominis_XP_665115 QKFDLNADF ribonucleotide reductase has been featured in almost 5,500 Cry_parvum_XP_627447 QKFDLNADF publications in a variety of felds, such as biochemistry, Cry_muris_XP_002140093 QIFNLDAEF The_annulata_XP_954052 QKFSTDLEF molecular biology, oncology, cell biology, and chemistry. As The_parva_XP_766246 QKFSTDLEF such, there is a wealth of information regarding this protein. R2_e1 only : * : :* The literature is replete with studies of RNR inhibitor use in R2_e2 and R2_e1 : . :* the control of cancer and of human pathogens. As detailed earlier, RNR has also received attention for its potential Figure 3. RNR subunit R2 C-terminus. A) Sequence logo use as a target to control the apicomplexan parasite P. representation of human and apicomplexan R2_e1 and falciparum. RNR inhibitors have already been shown to apicomplexan R2_e2 terminal residues. F1 and F7 are crucial have some antimalarial activity. Examples include RNA phenylalanine residues that interact with the R1 subunit. antisense oligonucleotide inhibitors (Chakrabarti et al., The numbering of these residues follows (Fisher et al., 1993) and radical scavengers such as the substituted/ 1993), while subsequent authors appear to have reversed modifed benzohydroxamic acids, specifcally the vicinal the order (Pellegrini et al., 2000; Pender et al., 2001; Gao dihydroxybenzohydroxamates (Holland et al., 1998). et al., 2002; Cooperman, 2003). Created with WebLogo 3 Knowledge of protein structure and localization can (Schneider and Stephens, 1990; Crooks et al., 2004). B) greatly aid in the design of chemotherapeutic drugs. For Terminal residues for human and apicomplexan R2_e1 and example, it is essential to determine if the region being apicomplexan R2_e2 from which the logos were derived. targeted is exposed on the protein's surface, whether Data derived from Munro et al. (submitted). Plasmodium and it is functional, and whether the residues surrounding human sequences are underlined for comparative purposes the target region in its native conformation are similarly (see text). Amino acid substitutions: . = semiconserved, : = conserved (Durand et al., 2008). While the structure of the conserved, * = identical. 16 Munro and Silva apicomplexan-specifc R2 subunit is not known, structure of suited for chemotherapeutical intervention, since it is the the orthodox R2_e1 subunit from (2O1Z) intraerythrocytic stages of Plasmodium that cause clinical and P. yoelii (2P1I) are deposited on the RCSB Protein Data symptoms (Yeh and Altman, 2006). Additionally, human Bank (www.pdb.org; Berman et al., 2000). RBCs are not nucleated, thus precluding an alternative Advances have been made in the production of means for the parasite to exploit host RNR (Rubin et al., apicomplexan recombinant proteins, a process that has 1993). been historically hampered by the A+T-biased nature of the The utility of RNR as an antimalarial/antipathogen target plasmodial genome and uncommon codon usage (Weber, depends upon the ability to specifcally target the pathogen's 1987; Anonymous, 2006; Brombacher 2006). However, RNR and not that of the host, as RNR is essential to both see Vedadi et al. (Vedadi et al., 2007) who found E. coli species. R1 and R2 subunits are highly conserved between to effectively produce apicomplexan proteins on a large- prokaryotes and eukaryotes in the regions containing the scale basis. Codon optimization (Hedfalk et al., 2008) functionally important residues (Chakrabarti et al., 1993; and codon harmonization (Hillier et al., 2005; Angov et al., Roshick et al., 2000; Voegtli et al., 2001; Högbom et al., 2008; Chowdhury et al., 2009) have been used to improve 2004; Högbom, 2010); however, they differ in the N- and expression of Plasmodium proteins in E. coli. Further C-terminal sequences (Ingram and Kinnaird, 1999). Novel advances have been made in the area of the use of a RNRs are additional potential targets for new drugs, phylogenetically similar, or “pseudoparasite”, expression especially if they provide distinct differences between host system (Fernádez-Robledo and Vasta, 2010). Furthermore, and parasite sequences. The necessity for target specifcity in vitro protocols for P. falciparum are established, although to avoid side, or non-target, effects in humans cannot in vivo models are based on the murine Plasmodium be overstated. The use of antisense oligonucleotides in species P. berghei, P. chabaudi, P. vinkei, and P. yoelii, and the control of some cancers has shown that the drugs in not those that parasitize humans (Fidock et al., 2004). question have favorable toxicity profles, in part because To a large degree, the utility of RNR as an antimalarial of their ability to specifcally target segments of RNA chemotherapeutic target is dependent upon the timing of (Davies et al., 2003). The unorthodox R2_e2 apicomplexan the protein's expression. In mammals and yeast, the large subunit provides a distinct and additional opportunity for subunit R1 has a half-life of 24 hours and is maintained specifc drug targeting. One example is the C-terminus of at a constant level throughout a cell's life cycle, while the the R2 subunit, which differs considerably between the small subunit R2 has a half-life of around 3 hours and its human R2_e1 subunits and both the R2_e1 and R2_e2 expression is restricted from the S-phase through to late apicomplexan subunits (Figures 3A and 3B). This difference mitosis, at which time it is rapidly degraded (Eriksson et al., in the C-terminal sequences of the R2 subunits between 1984; Engström et al., 1985; Björklund et al., 1990; Elledge apicomplexans and their hosts can be ideally targeted by et al., 1992). The non-canonical human p53R2 protein is chemotherapeutic means (Rubin et al., 1993; Fisher et al., expressed during periods of DNA repair (Håkansson et al., 1995; Ingram and Kinnaird, 1999). 2006). In mammalian cells, it has been demonstrated that it Finally, it is worth noting that Plasmodium-infected is the presence or absence of the R2 protein that regulates erythrocytes demonstrate an increase in cell membrane RNR activity (Chabes and Thelander, 2000). In contrast to permeability (Baumeister et al., 2011). In vitro uptake of this, in S. cerevisiae it is the R1 subunit whose transcription small pieces of RNA becomes less of an issue as RBCs that is increased during S-phase, thus controlling RNR activity are infected with malaria exhibit an enhanced and selective (Ortigosa et al., 2006). The utility of RNR inhibition in the uptake of such molecules in comparison to non-infected control of certain cancers has relied in part on the fact that RBCs (Rapaport et al., 1992), a difference attributed to RNR is most needed when cells are rapidly proliferating, the presence of a parasitophorous ducts in infected RBCs rendering cancerous cells particularly vulnerable to (Pouvelle et al., 1991). RNR inhibition (Smith and Karp, 2003). In P. falciparum, ribonucleotide biosynthesis begins as early as the ring Summary and early trophozoite stage; however, deoxyribonucleotide Extensive research has already established that RNR metabolism occurs later, with both R1 and R2_e1 subunit has potential as an antimalarial drug. What is particularly transcripts detected in the red blood cells (RBCs) at 10 appealing about RNR inhibition as a means of controlling hours post RBC infection and peaking 31 to 33 hours Apicomplexa is the potential control of not one, but two, post-infection, which coincides with the late trophozoite/ copies of the R2 subunit, both of which are distinct from that early schizont stage (Chakrabarti et al., 1993; Bozdech et of the host. Additionally, in the case of the malarial parasite al., 2003; Bozdech and Ginsburg, 2004). Comprehensive Plasmodium, RNR expression occurs from the sporozoite expression studies in P. falciparum show that the expression through the gametocyte life cycle stage, offering multiple profle of R2_e2, albeit less intense, matches that of the opportunities for chemotherapeutic targeting. This may well other to subunits (Bozdech et al., 2003; Llinás et al., 2006). be the case for other Apicomplexa parasites that undergo The timing of RNR expression is not surprising, since it is rapid clonal expansion in the host. during the intraerythrocytic stages that the malarial parasite Of the eight established methods of RNR inhibition undergoes logarithmic growth and requires RNR for DNA discussed, RNAi seems the least promising in terms of synthesis (Yeh and Altman, 2006). Additionally, expression controlling apicomplexan parasites as the necessary of PfR4 (R2_e2) has been detected in the sporozoite and enzymes appear to be lacking. Ribozyme approaches have gametocyte stages of the parasite life cycle (Bracchi-Ricard been successfully implemented in Plasmodium; however et al., 2005), and in the case of P. yoelii, both R1 and R2_ their use against RNR has not yet been demonstrated. In e1 transcripts were detected in the liver stage of infection contrast, substrate analogs and allosteric effector analogs (Nivez et al., 2000). These expression profles are well- have been effectively used to inhibit RNR, but their use Ribonucleotide Reductase as a Target to Control Apicomplexan Diseases 17 in Plasmodium has yet to be demonstrated. Antisense Aurelian, L., and Smith, C.C. (2000). Herpes simplex virus oligonucleotide inhibitors, dimerization inhibitors, radical type 2 growth and latency reactivation by cocultivation are scavengers, and iron chelators have all been successfully inhibited with antisense oligonucleotides complementary used to target RNR in Plasmodium. Ribozymes, antisense to the translation initiation site of the large subunit of oligonucleotide inhibitors, and dimerization inhibitors show ribonucleotide reductase (RR1). Antisense Nucleic Acid the most promise in terms of future anti-apicomplexan drug Drug Dev 10, 77-85. development. On the other hand, resistance to some radical Baker, B.F., and Monia, B.P. (1999). Novel mechanisms scavengers and iron chelators has been established, they for antisense-mediated regulation of gene expression. can not be use to selectively target a peptide, and the fact Biochim Biophys Acta 1489, 3-18. that the effects of iron chelators are generally reversible, Banach, M.J., and Williams, G.A. (2000). Purtscher makes them less appealing prospects. retinopathy and necrotizing vasculitis with gemcitabine The difference in sequence similarity between the therapy. Arch Ophthalmol 118, 726-727. parasite and human R2_e1 subunits is considerable, Barker, R.H., Metelev, V., Coakley, A., and Zamecnik, and the difference is even more accentuated when the P. (1998). Plasmodium falciparum: Effect of chemical parasite's R2_e2 subunit is considered (Munro et al., structure on effcacy and specifcity of antisense submitted). Assuming that this protein is essential for RNR oligonucleotides against malaria in vitro. Exp Parasitol function, ribozymes, antisense oligonucleotide inhibitors, 88, 51-59. and dimerization inhibitors can all be optimized for target Barker, R.H., Metelev, V., Rapaport, E., and Zamecnik, P. specifcity and thus used to take advantage of the unique (1996). Inhibition of Plasmodium falciparum malaria using R2_e2 protein. The fact that the R2_e2 gene is present antisense oligodeoxynucleotides. Proc Natl Acad Sci Unit in several apicomplexan genera, each of which contains States Am 93, 514-518. species of signifcant health and socio-economic impact, Baum, J., Papenfuss, A.T., Mair, G.R., Janse, C.J., holds promise that the results of any research would be Vlachou, D., Waters, A.P., Cowman, A.F., Crabb, B.S., translatable to several very important diseases. and de Koning-Ward, T.F. (2009). Molecular genetics and comparative genomics reveal RNAi is not functional in References malaria parasites. Nucleic Acids Res 37, 3788-3798. Aboul-Fadl, T. (2005). Antisense oligonucleotides: The state Baumeister, S., Wiesner, J., Reichenberg, A., Hintz, M., of the art. Curr Med Chem 12, 2193-2214. Bietz, S., Harb, O.S., Roos, D.S., Kordes, M., Friesen, Achenbach, T.V., Brunner, B., and Heermeier, K. (2003). J., Matuschewski, K., et al. (2011). Fosmidomycin uptake Oligonucleotide-based knockdown technologies: into Plasmodium and Babesia-infected erythrocytes is Antisense versus RNA interference. ChemBioChem 4, facilitated by parasite-induced new permeability pathways. 928-935. PLoS One 6, e19334. Adl, S.M., Simpson, A.G.B., Farmer, M.A., Andersen, R.A., Berman, H.M., Westbrook, J., Feng, Z., Gilliland, G., Bhat, Anderson, O.R., Barta, J.R., Bowser, S.S., Brugerolle, T.N., Weissig, H., Shindyalov, I.N., and Bourne, P.E. G., Fensome, R.A., Fredericq, S., et al. (2005). The new (2000). The Protein Data Bank. Nucleic Acids Res 28, higher level classifcation of eukaryotes with emphasis on 235-242. the of . J Eukaryot Microbiol 52, 399- Bianchi, V., Borella, S., Calderazzo, F., Ferraro, P., Chieco 451. Bianchi, L., and Reichard, P. (1994). Inhibition of Akiyoshi, D.E., Balakrishnan, R., Huettinger, C., Widmer, ribonucleotide reductase by 2'-substituted deoxycytidine G., and Tzipori, S. (2002). Molecular characterization of analogs: Possible application in AIDS treatment. Proc ribonucleotide reductase from Cryptosporidium parvum. Natl Acad Sci Unit States Am 91, 8403-8407. DNA Sequence 13, 167-172. Björklund, S., Skog, S., Tribukait, B., and Thelander, L. Alonso, P.L., Djimde, A., Kremsner, P., Magill, A., Milman, (1990). S-phase-specifc expression of mammalian J., Nájera, J., Plowe, C.V., Rabinovich, R., Wells, T., ribonucleotide reductase R1 and R2 subunit mRNAs. and Yeung, S. (2011). A research agenda for malaria Biochemistry 29, 5452-5458. eradication: Drugs. PLoS Medicine 8, e1000402. Blackman, M.J. (2003). RNAi in protozoan parasites: What Anderson, T. (2009). Mapping the spread of malaria drug hope for the Apicomplexa? 154, 177-180. resistance. PLoS Medicine 6, e1000054. Blanchard, J.L., and Hicks, J.S. (1999). The non- Andersson, C.S., and Högbom, M. (2009). A Mycobacterium photosynthetic plastid in malarial parasites and other tuberculosis ligand-binding Mn/Fe protein reveals a new apicomplexans is derived from outside the green plastid cofactor in a remodeled R2-protein scaffold. Proc Natl lineage. J Eukaryot Microbiol 46, 367-375. Acad Sci Unit States Am 106, 5633-5638. Bledsoe, G.H. (2005). Malaria primer for clinicians in the Angov, E., Hillier, C.J., Kincaid, R.L., and Lyon, J.A. United States. South Med J 98, 1197-1204; quiz 1205, (2008). Heterologous protein expression is enhanced by 1230. harmonizing the codon usage frequencies of the target Booden, T., and Hull, R.W. (1973). Nucleic acid precursor gene with those of the expression host. PLoS One 3, synthesis by parasitizing e2189. erythrocytes. Exp Parasitol 34, 220-228. Anonymous (2006). Crystal ball. Parasite Immunol 28, 235- Bourdon, A., Minai, L., Serre, V., Jais, J.-P., Sarzi, E., Aubert, 269. S., Chrétien, D., De Lonlay, P., Paquis-Flucklinger, V., Aravind, L., Iyer, L.M., Wellems, T.E., and Miller, L.H. (2003). Arakawa, H., et al. (2007). Mutation of RRM2B, encoding Plasmodium biology: Genomic gleanings. Cell 115, 771- p53-controlled ribonucleotide reductase (p53R2), causes 785. severe mitochondrial DNA depletion. Nat Genet 39, 776- 780. 18 Munro and Silva

Bozdech, Z., and Ginsburg, H. (2004). Antioxidant defense in Chakrabarti, D., Schuster, S.M., and Chakrabarti, R. Plasmodium falciparum--data mining of the transcriptome. (1993). Cloning and characterization of subunit genes of Malaria Journal 3, 23. ribonucleotide reductase, a cell-cycle-regulated enzyme, Bozdech, Z., Llinás, M., Pulliam, B.L., Wong, E.D., Zhu, from Plasmodium falciparum. Proc Natl Acad Sci Unit J., and DeRisi, J.L. (2003). The transcriptome of the States Am 90, 12020-12024. intraerythrocytic developmental cycle of Plasmodium Chan, J.H.P., Lim, S., and Wong, W.S.F. (2006). Antisense falciparum. PLoS Biol 1, E5. oligonucleotides: From design to therapeutic application. Bracchi-Ricard, V., Moe, D., and Chakrabarti, D. (2005). Clin Exp Pharmacol Physiol 33, 533-540. Two Plasmodium falciparum ribonucleotide reductase Chaudhary, K., Darling, J.A., Fohl, L.M., Sullivan, W.J., small subunits, PfR2 and PfR4, interact with each other Donald, R.G.K., Pfefferkorn, E.R., Ullman, B., and Roos, and are components of the in vivo enzyme complex. J Mol D.S. (2004). Purine salvage pathways in the apicomplexan Biol 347, 749-758. parasite . J Biol Chem 279, 31221- Brombacher , F. (2006). Crystal ball. Parasite Immunol 28, 31227. 235-269. Chen, S., Zhou, B., He, F., and Yen, Y. (2000). Inhibition Brown, A.E., and Catteruccia, F. (2006). Toward silencing of human cancer cell growth by inducible expression the burden of malaria: Progress and prospects for RNAi- of human ribonucleotide reductase antisense cDNA. based approaches. BioTechniques Suppl, 38-44. Antisense Nucleic Acid Drug Dev 10, 111-116. Brown, N.C., and Reichard, P. (1969). Role of effector Choi, K.S., Lee, T.H., and Jung, M.H. (2003). Ribozyme- binding in allosteric control of ribonucleoside diphosphate mediated cleavage of the human survivin mRNA and reductase. J Mol Biol 46, 39-55. inhibition of antiapoptotic function of survivin in MCF-7 Bustamante, C., Batista, C.N., and Zalis, M. (2009). cells. Cancer Gene Ther 10, 87-95. Molecular and biological aspects of antimalarial resistance Chowdhury, D.R., Angov, E., Kariuki, T., and Kumar, N. in Plasmodium falciparum and Plasmodium vivax. Curr (2009). A potent malaria transmission blocking vaccine Drug Targets 10, 279-290. based on codon harmonized full length Pfs48/45 Cao, M.-Y., Lee, Y., Feng, N.-P., Xiong, K., Jin, H., Wang, expressed in Escherichia coli. PLoS One 4, e6352. M., Vassilakos, A., Viau, S., Wright, J.A., and Young, A.H. Chrubasik, C., and Jacobson, R.L. (2010). The development (2003). Adenovirus-mediated ribonucleotide reductase of artemisinin resistance in malaria: Reasons and R1 gene therapy of human colon adenocarcinoma. Clin solutions. Phytother Res 24, 1104-1106. Cancer Res 9, 4553-4561. Citti, L., and Rainaldi, G. (2005). Synthetic hammerhead Carvalho, T.G., and Ménard, R. (2005). Manipulating the ribozymes as therapeutic tools to control disease genes. Plasmodium genome. Curr Issues Mol Biol 7, 39-55. Curr Gene Ther 5, 11-24. Cassera, M.B., Hazleton, K.Z., Riegelhaupt, P.M., Merino, Climent, I., Sjöberg, B.-M., and Huang, C.Y. (1991). E.F., Luo, M., Akabas, M.H., and Schramm, V.L. (2008). Carboxyl-terminal peptides as probes for Escherichia Erythrocytic adenosine monophosphate as an alternative coli ribonucleotide reductase subunit interaction: Kinetic purine source in Plasmodium falciparum. J Biol Chem analysis of inhibition studies. Biochemistry 30, 5164- 283, 32889-32899. 5171. Cerqueira, N.M.F.S.A., Fernandes, P.A., and Ramos, M.J. Climent, I., Sjöberg, B.-M., and Huang, C.Y. (1992). Site- (2007). Ribonucleotide reductase: A critical enzyme for directed mutagenesis and deletion of the carboxyl cancer chemotherapy and antiviral agents. Recent Pat terminus of Escherichia coli ribonucleotide reductase Anti-Canc 2, 11-29. protein R2. Effects on catalytic activity and subunit Cerqueira, N.M.F.S.A., Pereira, S., Fernandes, P.A., and interaction. Biochemistry 31, 4801-4807. Ramos, M.J. (2005). Overview of ribonucleotide reductase Cohen, E.A., Gaudreau, P., Brazeau, P., and Langelier, Y. inhibitors: An appealing target in anti-tumour therapy. Curr (1986). Specifc inhibition of herpesvirus ribonucleotide Med Chem 12, 1283-1294. reductase by a nonapeptide derived from the carboxy Cerutti, H., and Casas-Mollano, J.A. (2006). On the origin terminus of subunit 2. Nature 321, 441-443. and functions of RNA-mediated silencing: From protists Coombs, G.H. (1999). Biochemical peculiarities and drug to man. Curr Genet 50, 81-99. targets in Cryptosporidium parvum: Lessons from other Chabes, A., Domkin, V., Larsson, G., Liu, A., Graslund, coccidian parasites. Parasitol Today 15, 333-338. A., Wijmenga, S., and Thelander, L. (2000). Yeast Cooperman, B.S. (2003). Oligopeptide inhibition of class I ribonucleotide reductase has a heterodimeric iron-radical- ribonucleotide reductases. Biopolymers 71, 117-131. containing subunit. Proc Natl Acad Sci Unit States Am 97, Corso, P.S., Kramer, M.H., Blair, K.A., Addiss, D.G., Davis, 2474-2479. J.P., and Haddix, A.C. (2003). Cost of illness in the Chabes, A., Georgieva, B.P., Domkin, V., Zhao, X., Rothstein, 1993 waterborne Cryptosporidium outbreak, Milwaukee, R., and Thelander, L. (2003). Survival of DNA damage in Wisconsin. Emerg Infect Dis 9, 426-431. yeast directly depends on increased dNTP levels allowed Cory, J.G. (1988). Ribonucleotide reductase as a by relaxed feedback inhibition of ribonucleotide reductase. chemotherapeutic target. Adv Enzyme Regul 27, 437- Cell 112, 391-401. 455. Chabes, A., and Thelander, L. (2000). Controlled protein Cory, J.G., and Mansell, M.M. (1975). Studies on mammalian degradation regulates ribonucleotide reductase activity in ribonucleotide reductase inhibition by pyridoxal phosphate proliferating mammalian cells during the normal cell cycle and the dialdehyde derivatives of adenosine, adenosine and in response to DNA damage and replication blocks. J 5'-monophosphate, and adenosine 5'-triphosphate. Canc Biol Chem 275, 17747-17753. Res 35, 390-396. Ribonucleotide Reductase as a Target to Control Apicomplexan Diseases 19

Cosentino, G., Lavallée, P., Rakhit, S., Plante, R., Gaudette, Dharia, N.V., Bright, A.T., Westenberger, S.J., Barnes, Y., Lawetz, C., Whitehead, P.W., Duceppe, J.S., Lépine- S.W., Batalov, S., Kuhen, K., Borboa, R., Federe, G.C., Frenette, C., and Dansereau, N. (1991). Specifc inhibition McClean, C.M., Vinetz, J.M., et al. (2010). Whole- of ribonucleotide reductases by peptides corresponding genome sequencing and microarray analysis of ex vivo to the C-terminal of their second subunit. Biochem Cell Plasmodium vivax reveal selective pressure on putative Biol 69, 79-83. drug resistance genes. Proc Natl Acad Sci Unit States Am Coulson, R.M.R., Hall, N., and Ouzounis, C.A. (2004). 107, 20045-20050. Comparative genomics of transcriptional control in the Dondorp, A.M., Yeung, S., White, L., Nguon, C., Day, N.P.J., human malaria parasite Plasmodium falciparum. Genome Socheat, D., and von Seidlein, L. (2010). Artemisinin Res 14, 1548-1554. resistance: Current status and scenarios for containment. Cox-Singh, J., Davis, T.M.E., Lee, K.-S., Shamsul, S.S.G., Nat Rev Microbiol 8, 272-280. Matusop, A., Ratnam, S., Rahman, H.A., Conway, D.J., Doolan, D.L., Aguiar, J.C., Weiss, W.R., Sette, A., Felgner, and Singh, B. (2008). Plasmodium knowlesi malaria P.L., Regis, D.P., Quinones-Casas, P., Yates, J.R., Blair, in humans is widely distributed and potentially life P.L., Richie, T.L., et al. (2003). Utilization of genomic threatening. Clin Infect Dis 46, 165-171. sequence information to develop malaria vaccines. J Exp Croft, S.L. (1997). The current status of antiparasite Biol 206, 3789-3802. chemotherapy. Parasitology 114, S3-S15. Dormeyer, M., Schöneck, R., Dittmar, G.A., and Krauth- Crona, M., Furrer, E., Torrents, E., Edgell, D.R., and Sjöberg, Siegel, R.L. (1997). Cloning, sequencing and expression B.-M. (2010). Subunit and small-molecule interaction of of ribonucleotide reductase R2 from Trypanosoma brucei. ribonucleotide reductases via surface plasmon resonance FEBS Lett 414, 449-453. biosensor analyses. Protein Eng Des Sel 23, 633-641. Downie, M.J., Kirk, K., and Mamoun, C.B. (2008). Purine Crooke, A., Diez, A., Mason, P.J., and Bautista, J.M. salvage pathways in the intraerythrocytic malaria parasite (2006). Transient silencing of Plasmodium falciparum Plasmodium falciparum. Eukaryot Cell 7, 1231-1237. bifunctional glucose-6-phosphate dehydrogenase- Durand, P.M., Naidoo, K., and Coetzer, T.L. (2008). 6-phosphogluconolactonase. FEBS J 273, 1537-1546. Evolutionary patterning: A novel approach to the Crooks, G.E., Hon, G., Chandonia, J.-M., and Brenner, S.E. identifcation of potential drug target sites in Plasmodium (2004). WebLogo: a sequence logo generator. Genome falciparum. PLoS One 3, e3685. Research 14, 1188-1190. Dutia, B.M., Frame, M.C., Subak-Sharpe, J.H., Clark, Dahl, E.L., and Rosenthal, P.J. (2008). Apicoplast W.N., and Marsden, H.S. (1986). Specifc inhibition translation, transcription and genome replication: Targets of herpesvirus ribonucleotide reductase by synthetic for antimalarial antibiotics. Trends Parasitol 24, 279-284. peptides. Nature 321, 439-441. Dahl, E.L., Shock, J.L., Shenai, B.R., Gut, J., DeRisi, J.L., Ekanem, J. (2001). Ribonucleotide reductase as target for and Rosenthal, P.J. (2006). Tetracyclines specifcally drug discovery against African sleeping sickness. NISEB target the apicoplast of the malaria parasite Plasmodium Journal 1, 287-292. falciparum. Antimicrob Agents Chemother 50, 3124- El Bissati, K., Zufferey, R., Witola, W.H., Carter, N.S., Ullman, 3131. B., and Ben Mamoun, C. (2006). The plasma membrane Dasaradhi, P.V.N., Mohmmed, A., Kumar, A., Hossain, M.J., permease PfNT1 is essential for purine salvage in the Bhatnagar, R.K., Chauhan, V.S., and Malhotra, P. (2005). human malaria parasite Plasmodium falciparum. Proc A role of falcipain-2, principal cysteine proteases of Natl Acad Sci Unit States Am 103, 9286-9291. Plasmodium falciparum in merozoite egression. Biochem Elledge, S.J., and Davis, R.W. (1990). Two genes Biophys Res Comm 336, 1062-1068. differentially regulated in the cell cycle and by DNA- Davies, A.M., Gandara, D.R., Lara, P.N., Mack, P.C., damaging agents encode alternative regulatory subunits Lau, D.H.M., and Gumerlock, P.H. (2003). Antisense of ribonucleotide reductase. Gene Dev 4, 740-751. oligonucleotides in the treatment of non-small-cell lung Elledge, S.J., Zhou, Z., and Allen, J.B. (1992). Ribonucleotide cancer. Clin Lung Canc 4 Suppl 2, S68-73. reductase: Regulation, regulation, regulation. Trends Davis, R., Thelander, M., Mann, G.J., Behravan, G., Soucy, Biochem Sci 17, 119-123. F., Beaulieu, P., Lavallée, P., Gräslund, A., and Thelander, Engström, Y., Eriksson, S., Jildevik, I., Skog, S., Thelander, L. (1994). Purifcation, characterization, and localization L., and Tribukait, B. (1985). Cell cycle-dependent of subunit interaction area of recombinant mouse expression of mammalian ribonucleotide reductase. ribonucleotide reductase R1 subunit. J Biol Chem 269, Differential regulation of the two subunits. J Biol Chem 23171-23176. 260, 9114-9116. de Koning, H.P., Bridges, D.J., and Burchmore, R.J.S. Enserink, M. (2010). Malaria's drug miracle in danger. (2005). Purine and pyrimidine transport in pathogenic Science 328, 844-846. : From biology to therapy. FEMS Microbiol Rev Ericsson, D.J., Nurbo, J., Muthas, D., Hertzberg, K., 29, 987-1020. Lindeberg, G., Karlén, A., and Unge, T. (2010). Desai, A.A., Schilsky, R.L., Young, A., Janisch, L., Stadler, Identifcation of small peptides mimicking the R2 W.M., Vogelzang, N.J., Cadden, S., Wright, J.A., and C-terminus of Mycobacterium tuberculosis ribonucleotide Ratain, M.J. (2005). A phase I study of antisense reductase. J Pept Sci 16, 159-164. oligonucleotide GTI-2040 given by continuous intravenous Eriksson, S., Gräslund, A., Skog, S., Thelander, L., and infusion in patients with advanced solid tumors. Ann Oncol Tribukait, B. (1984). Cell cycle-dependent regulation 16, 958-965. of mammalian ribonucleotide reductase. The S phase- correlated increase in subunit M2 is regulated by de novo protein synthesis. J Biol Chem 259, 11695-11700. 20 Munro and Silva

Eriksson, S., Thelander, L., and Åkerman, M. (1979). Gardner, M.J., Hall, N., Fung, E., White, O., Berriman, Allosteric regulation of calf thymus ribonucleoside M., Hyman, R.W., Carlton, J.M., Pain, A., Nelson, K.E., diphosphate reductase. Biochemistry 18, 2948-2952. Bowman, S., et al. (2002). Genome sequence of the Fairman, J.W., Wijerathna, S.R., Ahmad, M.F., Xu, H., human malaria parasite Plasmodium falciparum. Nature Nakano, R., Jha, S., Prendergast, J., Welin, R.M., 419, 498-511. Flodin, S., Roos, A., et al. (2011). Structural basis for Gaudreau, P., Brazeau, P., Richer, M., Cormier, J., Langlois, allosteric regulation of human ribonucleotide reductase D., and Langelier, Y. (1992). Structure-function studies of by nucleotide-induced oligomerization. Nat Struct Biol 18, peptides inhibiting the ribonucleotide reductase activity of 316-322. herpes simplex virus type I. J Med Chem 35, 346-350. Fan, H., Huang, A., Villegas, C., and Wright, J.A. (1997). The Gaudreau, P., Michaud, J., Cohen, E.A., Langelier, Y., and R1 component of mammalian ribonucleotide reductase Brazeau, P. (1987). Structure-activity studies on synthetic has malignancy-suppressing activity as demonstrated by peptides inhibiting herpes simplex virus ribonucleotide gene transfer experiments. Proc Natl Acad Sci Unit States reductase. J Biol Chem 262, 12413-12416. Am 94, 13181-13186. Gaudreau, P., Paradis, H., Langelier, Y., and Brazeau, P. Fan, H., Villegas, C., Huang, A., and Wright, J.A. (1998). (1990). Synthesis and inhibitory potency of peptides The mammalian ribonucleotide reductase R2 component corresponding to the subunit 2 C-terminal region of cooperates with a variety of oncogenes in mechanisms of herpes virus ribonucleotide reductases. J Med Chem 33, cellular transformation. Canc Res 58, 1650-1653. 723-730. Fast, N.M., Kissinger, J.C., Roos, D.S., and Keeling, Ge, J., Perlstein, D.L., Nguyen, H.H., Bar, G., Griffn, R.G., P.J. (2001). Nuclear-encoded, plastid-targeted genes and Stubbe, J. (2001). Why multiple small subunits (Y2 suggest a single common origin for apicomplexan and and Y4) for yeast ribonucleotide reductase? Toward dinofagellate plastids. Mol Biol Evol 18, 418-426. understanding the role of Y4. Proc Natl Acad Sci Unit Fernádez-Robledo, J.A., and Vasta, G.R. (2010). Production States Am 98, 10067-10072. of recombinant proteins from protozoan parasites. Trends Gissot, M., Briquet, S., Refour, P., Boschet, C., and Vaquero, Parasitol 26, 244-254. C. (2005). PfMyb1, a Plasmodium falciparum transcription Fidock, D.A. (2010). Drug discovery: Priming the antimalarial factor, is required for intra-erythrocytic growth and controls pipeline. Nature 465, 297-298. key genes for cell cycle regulation. J Mol Biol 346, 29- Fidock, D.A., Rosenthal, P.J., Croft, S.L., Brun, R., and 42. Nwaka, S. (2004). Antimalarial drug discovery: Effcacy Gon, S., Camara, J.E., Klungsøyr, H.K., Crooke, E., models for compound screening. Nat Rev Drug Discov Skarstad, K., and Beckwith, J. (2006). A novel regulatory 3, 509-520. mechanism couples deoxyribonucleotide synthesis and Fisher, A., Laub, P.B., and Cooperman, B.S. (1995). NMR DNA replication in Escherichia coli. EMBO J 25, 1137- structure of an inhibitory R2 C-terminal peptide bound to 1147. mouse ribonucleotide reductase R1 subunit. Nat Struct Greenwood, B., and Mutabingwa, T. (2002). Malaria in 2002. Biol 2, 951-955. Nature 415, 670-672. Fisher, A., Yang, F.D., Rubin, H., and Cooperman, B.S. Gregson, A., and Plowe, C.V. (2005). Mechanisms of (1993). R2 C-terminal peptide inhibition of mammalian resistance of malaria parasites to antifolates. Pharmacol and yeast ribonucleotide reductase. J Med Chem 36, Rev 57, 117-145. 3859-3862. Grimm, D., and Kay, M.A. (2007). Therapeutic application Flores, M.V., Atkins, D., Wade, D., O'Sullivan, W.J., and of RNAi: Is mRNA targeting fnally ready for prime time? J Stewart, T.S. (1997). Inhibition of Plasmodium falciparum Clin Invest 117, 3633-3641. proliferation in vitro by ribozymes. J Biol Chem 272, Guittet, O., Håkansson, P., Voevodskaya, N., Fridd, S., 16940-16945. Gräslund, A., Arakawa, H., Nakamura, Y., and Thelander, Fontecave, M. (1998). Ribonucleotide reductases and L. (2001). Mammalian p53R2 protein forms an active radical reactions. Cell Mol Life Sci 54, 684-695. ribonucleotide reductase in vitro with the R1 protein, Fontecave, M., Lepoivre, M., Elleingand, E., Gerez, C., and which is expressed both in resting cells in response to Guittet, O. (1998). Resveratrol, a remarkable inhibitor of DNA damage and in proliferating cells. J Biol Chem 276, ribonucleotide reductase. FEBS Lett 421, 277-279. 40647-40651. Foth, B.J., Ralph, S.A., Tonkin, C.J., Struck, N.S., Fraunholz, Guittet, O., Roy, B., and Lepoivre, M. (1999). Nitric oxide: A M., Roos, D.S., Cowman, A.F., and McFadden, G.I. radical molecule in quest of free radicals in proteins. Cell (2003). Dissecting apicoplast targeting in the malaria Mol Life Sci 55, 1054-1067. parasite Plasmodium falciparum. Science 299, 705-708. Gunasekera, A.M., Patankar, S., Schug, J., Eisen, G., Fu, Y., and Xiao, W. (2006). Identifcation and characterization Kissinger, J.C., Roos, D., and Wirth, D.F. (2004). of CRT10 as a novel regulator of Saccharomyces Widespread distribution of antisense transcripts in the cerevisiae ribonucleotide reductase genes. Nucleic Acids Plasmodium falciparum genome. Mol Biochem Parasitol Res 34, 1876-1883. 136, 35-42. Gao, Y., Liehr, S., and Cooperman, B.S. (2002). Affnity- Gwilt, P.R., and Tracewell, W.G. (1998). Pharmacokinetics driven selection of tripeptide inhibitors of ribonucleotide and pharmacodynamics of hydroxyurea. Clin reductase. Bioorg Med Chem Lett 12, 513-515. Pharmacokinet 34, 347-358. Gardiner, D.L., Holt, D.C., Thomas, E.A., Kemp, D.J., Håkansson, P., Hofer, A., and Thelander, L. (2006). and Trenholme, K.R. (2000). Inhibition of Plasmodium Regulation of mammalian ribonucleotide reduction and falciparum clag9 gene function by antisense RNA. Mol dNTP pools after DNA damage and in resting cells. J Biol Biochem Parasitol 110, 33-41. Chem 281, 7834-7841. Ribonucleotide Reductase as a Target to Control Apicomplexan Diseases 21

Harder, J. (1993). Ribonucleotide reductases and their Inselburg, J., and Banyal, H.S. (1984). Synthesis of DNA occurrence in microorganisms: A link to the RNA/DNA during the asexual cycle of Plasmodium falciparum in transition. FEMS Microbiol Rev 12, 273-292. culture. Mol Biochem Parasitol 10, 79-87. Harrington, J.A., and Spector, T. (1991). Human Janouškovec, J., Horák, A., Oborník, M., Lukeš, J., and ribonucleotide reductase. Activation and inhibition by Keeling, P.J. (2010). A common red algal origin of the analogs of ATP. Biochem Pharmacol 42, 759-763. apicomplexan, dinofagellate, and plastids. Haseloff, J., and Gerlach, W.L. (1988). Simple RNA enzymes Proc Natl Acad Sci Unit States Am 107, 10949-10954. with new and highly specifc endoribonuclease activities. Jeha, S., Gandhi, V., Chan, K.W., McDonald, L., Ramirez, I., Nature 334, 585-591. Madden, R., Rytting, M., Brandt, M., Keating, M., Plunkett, Hedfalk, K., Pettersson, N., Oberg, F., Hohmann, S., and W., et al. (2004). Clofarabine, a novel nucleoside analog, Gordon, E. (2008). Production, characterization and is active in pediatric patients with advanced leukemia. crystallization of the Plasmodium falciparum aquaporin. Blood 103, 784-789. Protein Expr Purif 59, 69-78. Jordan, A., Pontis, E., Atta, M., Krook, M., Gibert, I., Barbé, Herrick, J., and Sclavi, B. (2007). Ribonucleotide reductase J., and Reichard, P. (1994). A second class I ribonucleotide and the regulation of DNA replication: An old story and an reductase in Enterobacteriaceae: Characterization of the ancient heritage. Mol Microbiol 63, 22-34. Salmonella typhimurium enzyme. Proc Natl Acad Sci Unit Hillier, C.J., Ware, L.A., Barbosa, A., Angov, E., Lyon, States Am 91, 12892-12896. J.A., Heppner, D.G., and Lanar, D.E. (2005). Process Jordan, A., and Reichard, P. (1998). Ribonucleotide development and analysis of liver-stage antigen 1, reductases. Annu Rev Biochem 67, 71-98. a preerythrocyte-stage protein-based vaccine for Khan, A.U. (2006). Ribozyme: a clinical tool. Clin Chim Acta Plasmodium falciparum. Infect Immun 73, 2109-2115. 367, 20-27. Ho, S.P., Britton, D.H., Stone, B.A., Behrens, D.L., Leffet, Kolberg, M., Strand, K.R., Graff, P., and Andersson, L.M., Hobbs, F.W., Miller, J.A., and Trainor, G.L. (1996). K.K. (2004). Structure, function, and mechanism of Potent antisense oligonucleotides to the human multidrug ribonucleotide reductases. Biochim Biophys Acta 1699, resistance-1 mRNA are rationally selected by mapping 1-34. RNA-accessible sites with oligonucleotide libraries. Krakoff, I.H., Brown, N.C., and Reichard, P. (1968). Inhibition Nucleic Acids Res 24, 1901-1907. of ribonucleoside diphosphate reductase by hydroxyurea. Hodges, Y.K., Antholine, W.E., and Horwitz, L.D. (2004). Canc Res 28, 1559-1565. Effect on ribonucleotide reductase of novel lipophilic iron Krug, E.C., Marr, J.J., and Berens, R.L. (1989). Purine chelators: The desferri-exochelins. Biochem Biophys Res metabolism in Toxoplasma gondii. J Biol Chem 264, Comm 315, 595-598. 10601-10607. Hofer, A., Schmidt, P.P., Gräslund, A., and Thelander, L. Kumar, D., Viberg, J., Nilsson, A.K., and Chabes, A. (2010). (1997). Cloning and characterization of the R1 and R2 Highly mutagenic and severely imbalanced dNTP pools subunits of ribonucleotide reductase from Trypanosoma can escape detection by the S-phase checkpoint. Nucleic brucei. Proc Natl Acad Sci Unit States Am 94, 6959- Acids Res. 6964. Kumar, R., Adams, B., Oldenburg, A., Musiyenko, A., and Högbom, M. (2010). The manganese/iron-carboxylate Barik, S. (2002). Characterisation and expression of a PP1 proteins: What is what, where are they, and what can the serine/threonine protein phosphatase (PfPP1) from the sequences tell us? J Biol Inorg Chem 15, 339-349. malaria parasite, Plasmodium falciparum: Demonstration Högbom, M., Stenmark, P., Voevodskaya, N., McClarty, G., of its essential role using RNA interference. Malaria Gräslund, A., and Nordlund, P. (2004). The radical site Journal 1, 5. in chlamydial ribonucleotide reductase defnes a new R2 Kyes, S., Christodoulou, Z., Pinches, R., and Newbold, subclass. Science 305, 245-248. C. (2002). Stage-specifc merozoite surface protein 2 Holland, K.P., Elford, H.L., Bracchi-Ricard, V., Annis, C.G., antisense transcripts in Plasmodium falciparum. Mol Schuster, S.M., and Chakrabarti, D. (1998). Antimalarial Biochem Parasitol 123, 79-83. activities of polyhydroxyphenyl and hydroxamic acid Lane, J., Martin, T.A., and Jiang, W.G. (2010). Targeting derivatives. Antimicrob Agents Chemother 42, 2456- RhoC by way of ribozyme trangene in human breast 2458. cancer cells and its impact on cancer invasion. World J Huang, M., and Elledge, S.J. (1997). Identifcation of Oncol 1, 7. RNR4, encoding a second essential small subunit of Leander, B.S., and Keeling, P.J. (2003). Morphostasis in ribonucleotide reductase in Saccharomyces cerevisiae. evolution. Trends Ecol Evol 18, 395-402. Mol Cell Biol 17, 6105-6113. Lee, S.H., and Sinko, P.J. (2006). siRNA—Getting the Huthmacher, C., Hoppe, A., Bulik, S., and Holzhütter, message out. Eur J Pharm Sci 27, 401-410. H.-G. (2010). Antimalarial drug targets in Plasmodium Lee, Y., Vassilakos, A., Feng, N., Lam, V., Xie, H., Wang, falciparum predicted by stage-specifc metabolic network M., Jin, H., Xiong, K., Liu, C., Wright, J., et al. (2003). analysis. BMC Syst Biol 4, 120. GTI-2040, an antisense agent targeting the small subunit Ingemarson, R., and Thelander, L. (1996). A kinetic study component (R2) of human ribonucleotide reductase, on the infuence of nucleoside triphosphate effectors on shows potent antitumor activity against a variety of subunit interaction in mouse ribonucleotide reductase. tumors. Canc Res 63, 2802-2811. Biochemistry 35, 8603-8609. Lepoivre, M., Fieschi, F., Coves, J., Thelander, L., and Ingram, G.M., and Kinnaird, J.H. (1999). Ribonucleotide Fontecave, M. (1991). Inactivation of ribonucleotide reductase: A new target for antiparasite therapies. reductase by nitric oxide. Biochem Biophys Res Comm Parasitol Today 15, 338-342. 179, 442-448. 22 Munro and Silva

Levine, N.D. (1988). Progress in taxonomy of the McRobert, L., and McConkey, G.A. (2002). RNA interference apicomplexan protozoa. J Protozool 35, 518-520. (RNAi) inhibits growth of Plasmodium falciparum. Mol Lewin, A.S., and Hauswirth, W.W. (2001). Ribozyme gene Biochem Parasitol 119, 273-278. therapy: Applications for molecular medicine. Trends Mol Mdluli, K., and Spigelman, M. (2006). Novel targets for Med 7, 221-228. tuberculosis drug discovery. Curr Opin Pharmacol 6, 459- Li, N.-S., Frederiksen, J.K., Koo, S.C., Lu, J., Wilson, 467. T.J., Lilley, D.M.J., and Piccirilli, J.A. (2010). A general Militello, K.T., Patel, V., Chessler, A.-D., Fisher, J.K., Kasper, and effcient approach for the construction of RNA J.M., Gunasekera, A.M., and Wirth, D.F. (2005). RNA oligonucleotides containing a 5'-phosphorothiolate polymerase II synthesizes antisense RNA in Plasmodium linkage. Nucleic Acids Res, 1-16. falciparum. RNA 11, 365-370. Lin, A.L., and Elford, H.L. (1980). Adenosine deaminase Ming, X., Alam, M.R., Fisher, M., Yan, Y., Chen, X., and impairment and ribonucleotide reductase activity and Juliano, R.L. (2010). Intracellular delivery of an antisense levels in HeLa cells. J Biol Chem 255, 8523-8528. oligonucleotide via endocytosis of a G protein-coupled Lin, Z.P., Belcourt, M.F., Cory, J.G., and Sartorelli, A.C. (2004). receptor. Nucleic Acids Res. Stable suppression of the R2 subunit of ribonucleotide Mkulama, M.A.P., Chishimba, S., Sikalima, J., Rouse, P., reductase by R2-targeted short interference RNA Thuma, P.E., and Mharakurwa, S. (2008). Escalating sensitizes p53(-/-) HCT-116 colon cancer cells to DNA- Plasmodium falciparum antifolate drug resistance damaging agents and ribonucleotide reductase inhibitors. mutations in Macha, rural Zambia. Malaria Journal 7, 87. J Biol Chem 279, 27030-27038. Mohmmed, A., Dasaradhi, P.V.N., Bhatnagar, R.K., Chauhan, Liuzzi, M., Déziel, R., Moss, N., Beaulieu, P., Bonneau, A.M., V.S., and Malhotra, P. (2003). In vivo gene silencing in Bousquet, C., Chafouleas, J.G., Garneau, M., Jaramillo, —a mouse malaria model. Biochem J., and Krogsrud, R.L. (1994). A potent peptidomimetic Biophys Res Comm 309, 506-511. inhibitor of HSV ribonucleotide reductase with antiviral Morrison, D.A. (2009). Evolution of the Apicomplexa: Where activity in vivo. Nature 372, 695-698. are we now? Trends Parasitol 25, 375-382. Lizundia, R., Werling, D., Langsley, G., and Ralph, S.A. Moss, N., Déziel, R., Adams, J., Aubry, N., Bailey, M., Baillet, (2009). Theileria apicoplast as a target for chemotherapy. M., Beaulieu, P., DiMaio, J., Duceppe, J.S., Ferland, Antimicrob Agents Chemother 53, 1213-1217. J.M., et al. (1993). Inhibition of herpes simplex virus type Llinás, M., Bozdech, Z., Wong, E.D., Adai, A.T., and DeRisi, 1 ribonucleotide reductase by substituted tetrapeptide J.L. (2006). Comparative whole genome transcriptome derivatives. J Med Chem 36, 3005-3009. analysis of three Plasmodium falciparum strains. Nucleic Mu, J., Seydel, K.B., Bates, A., and Su, X.-z. (2010). Recent Acids Res 34, 1166-1173. progress in functional genomic research in Plasmodium Logan, D.T. (2011). Closing the circle on ribonucleotide falciparum. Curr Genom 11, 279-286. reductases. Nat Struct Biol 18, 251-253. Nayak, A., and Kohli, D. (2005). Ribozymes: The trans Lou, Z., and Zhang, X. (2010). Protein targets for structure- acting tools for RNA manipulation. Indian Journal of based anti-Mycobacterium tuberculosis drug discovery. Pharmaceutical Education 39, 92-96. Protein Cell 1, 435-442. Nguyen, H.H., Ge, J., Perlstein, D.L., and Stubbe, J. (1999). Lu, Y., Gu, J., Jin, D., Gao, Y., and Yuan, M. (2011). Inhibition Purifcation of ribonucleotide reductase subunits Y1, of telomerase activity by HDV ribozyme in cancers. J Exp Y2, Y3, and Y4 from yeast: Y4 plays a key role in diiron Clin Canc Res 30, 1. cluster assembly. Proc Natl Acad Sci Unit States Am 96, Lundin, D., Torrents, E., Poole, A.M., and Sjöberg, B.-M. 12339-12344. (2009). RNRdb, a curated database of the universal Nivez, M., Achbarou, A., Bienvenu, J.D., Mazier, D., enzyme family ribonucleotide reductase, reveals a Doerig, C., and Vaquero, C. (2000). A study of selected high level of misannotation in sequences deposited to Plasmodium yoelii messenger RNAs during hepatocyte GenBank. BMC Genomics 10, 589. infection. Mol Biochem Parasitol 111, 31-39. Lytton, S.D., Mester, B., Libman, J., Shanzer, A., and Nocentini, G. (1996). Ribonucleotide reductase inhibitors: Cabantchik, Z.I. (1994). Mode of action of iron (III) New strategies for cancer chemotherapy. Crit Rev Oncol chelators as antimalarials: II. Evidence for differential Hematol 22, 89-126. effects on parasite iron-dependent nucleic acid synthesis. Nocentini, G., Federici, F., Armellini, R., Franchetti, P., and Blood 84, 910-915. Barzi, A. (1990). Isolation of two cellular lines resistant Madrid, D.C., Ting, L.-M., Waller, K.L., Schramm, V.L., and to ribonucleotide reductase inhibitors to investigate the Kim, K. (2008). Plasmodium falciparum purine nucleoside inhibitory activity of 2,2'-bipyridyl-6-carbothioamide. Anti phosphorylase is critical for viability of malaria parasites. Canc Drugs 1, 171-177. J Biol Chem 283, 35899-35907. Noonpakdee, W., Pothikasikorn, J., Nimitsantiwong, W., and Malhotra, P., Dasaradhi, P.V.N., Kumar, A., Mohmmed, Wilairat, P. (2003). Inhibition of Plasmodium falciparum A., Agrawal, N., Bhatnagar, R.K., and Chauhan, V.S. proliferation in vitro by antisense oligodeoxynucleotides (2002). Double-stranded RNA-mediated gene silencing against malarial topoisomerase II. Biochem Biophys Res of cysteine proteases (falcipain-1 and -2) of Plasmodium Comm 302, 659-664. falciparum. Mol Microbiol 45, 1245-1254. Nordlund, P., and Eklund, H. (1993). Structure and function Mathews, C.K. (2006). DNA precursor metabolism and of the Escherichia coli ribonucleotide reductase protein genomic stability. FASEB J 20, 1300-1314. R2. J Mol Biol 232, 123-164. McFadden, G.I., and Roos, D.S. (1999). Apicomplexan Nordlund, P., and Reichard, P. (2006). Ribonucleotide plastids as drug targets. Trends Microbiol 7, 328-333. reductases. Annu Rev Biochem 75, 681-706. Ribonucleotide Reductase as a Target to Control Apicomplexan Diseases 23

Nordlund, P., Sjöberg, B.-M., and Eklund, H. (1990). Perez, M., Cerqueira, N., Fernandes, P., and Ramos, M. Three-dimensional structure of the free radical protein of (2010). Ribonucleotide reductase: A mechanistic portrait ribonucleotide reductase. Nature 345, 593-598. of substrate analogues inhibitors. Curr Med Chem 17, Norris, J.S., Hoel, B., Voeks, D., Maggouta, F., Dahm, M., 2854-2872. Pan, W., and Clawson, G. (2000). Design and testing of Perlstein, D.L., Ge, J., Ortigosa, A.D., Robblee, J.H., Zhang, ribozymes for cancer gene therapy. Adv Exp Med Biol Z., Huang, M., and Stubbe, J. (2005). The active form of 465, 293-301. the Saccharomyces cerevisiae ribonucleotide reductase Nosrati, M., Li, S., Bagheri, S., Ginzinger, D., Blackburn, small subunit is a heterodimer in vitro and in vivo. E.H., Debs, R.J., and Kashani-Sabet, M. (2004). Biochemistry 44, 15366-15377. Antitumor activity of systemically delivered ribozymes Pouvelle, B., Spiegel, R., Hsiao, L., Howard, R.J., Morris, targeting murine telomerase RNA. Clin Cancer Res 10, R.L., Thomas, A.P., and Taraschi, T.F. (1991). Direct 4983-4990. access to serum macromolecules by intraerythrocytic Nyholm, S., Mann, G.J., Johansson, A.G., Bergeron, malaria parasites. Nature 353, 73-75. R.J., Gräslund, A., and Thelander, L. (1993). Role of Pradines, B., Ramiandrasoa, F., Basco, L.K., Bricard, L., ribonucleotide reductase in inhibition of mammalian cell Kunesch, G., and Le Bras, J. (1996). In vitro activities growth by potent iron chelators. J Biol Chem 268, 26200- of novel catecholate siderophores against Plasmodium 26205. falciparum. Antimicrob Agents Chemother 40, 2094- Olliaro, P.L., and Yuthavong, Y. (1999). An overview of 2098. chemotherapeutic targets for antimalarial drug discovery. Prusty, D., Dar, A., Priya, R., Sharma, A., Dana, S., Pharmacol Therapeut 81, 91-110. Choudhury, N.R., Rao, N.S., and Dhar, S.K. (2010). Ortigosa, A.D., Hristova, D., Perlstein, D.L., Zhang, Z., Single-stranded DNA binding protein from human malarial Huang, M., and Stubbe, J. (2006). Determination of parasite Plasmodium falciparum is encoded in the nucleus the in vivo stoichiometry of tyrosyl radical per ββ' in and targeted to the apicoplast. Nucleic Acids Res. Saccharomyces cerevisiae ribonucleotide reductase. Puerta-Fernández, E., Romero-López, C., Barroso- Biochemistry 45, 12282-12294. delJesus, A., and Berzal-Herranz, A. (2003). Ribozymes: Padmanaban, G. (2003). Drug targets in malaria parasites. Recent advances in the development of RNA tools. FEMS Adv Biochem Eng Biotechnol 84, 123-141. Microbiol Rev 27, 75-97. Paradis, H., Gaudreau, P., Brazeau, P., and Langelier, Ralph, S., van Dooren, G., Waller, R., Crawford, M.J., Y. (1988). Mechanism of inhibition of herpes simplex Fraunholz, M., Foth, B.J., Tonkin, C., Roos, D., and virus (HSV) ribonucleotide reductase by a nonapeptide McFadden, G.I. (2004). Metabolic maps and functions of corresponding to the carboxyl terminus of its subunit 2. the Plasmodium falciparum apicoplast. Nat Rev Microbiol Specifc binding of a photoaffnity analog, [4'- azido-Phe6] 2, 203-216. HSV H2-6(6-15), to subunit 1. J Biol Chem 263, 16045- Ralph, S.A., D'Ombrain, M.C., and McFadden, G.I. (2001). 16050. The apicoplast as an antimalarial drug target. Drug Resist Parker, M.D., Hyde, R.J., Yao, S.Y., McRobert, L., Cass, C.E., Updates 4, 145-151. Young, J.D., McConkey, G.A., and Baldwin, S.A. (2000). Raman, J., and Balaram, H. (2004). Nucleotide metabolism Identifcation of a nucleoside/nucleobase transporter from in Plasmodium falciparum: Recent developments. Med Plasmodium falciparum, a novel target for anti-malarial Chem Rev Online 1, 465-473. chemotherapy. Biochem J 349, 67-75. Rapaport, E., Misiura, K., Agrawal, S., and Zamecnik, P. Patankar, S., Munasinghe, A., Shoaibi, A., Cummings, L.M., (1992). Antimalarial activities of oligodeoxynucleotide and Wirth, D.F. (2001). Serial analysis of gene expression phosphorothioates in chloroquine-resistant Plasmodium in Plasmodium falciparum reveals the global expression falciparum. Proc Natl Acad Sci Unit States Am 89, 8577- profle of erythrocytic stages and the presence of anti- 8580. sense transcripts in the malarial parasite. Mol Biol Cell Reichard, P. (1988). Interactions between deoxyribonucleotide 12, 3114-3125. and DNA synthesis. Annu Rev Biochem 57, 349-374. Pellegrini, M., Liehr, S., Fisher, A.L., Laub, P.B., Cooperman, Reichard, P. (1997). The evolution of ribonucleotide B.S., and Mierke, D.F. (2000). Structure-based optimization reduction. Trends Biochem Sci 22, 81-85. of peptide inhibitors of mammalian ribonucleotide Reichard, P. (2002). Ribonucleotide reductases: The reductase. Biochemistry 39, 12210-12215. evolution of allosteric regulation. Arch Biochem Biophys Pender, B.A., Wu, X., Axelsen, P.H., and Cooperman, B.S. 397, 149-155. (2001). Toward a rational design of peptide inhibitors Reichard, P. (2010). Ribonucleotide reductases: Substrate of ribonucleotide reductase: Structure—function and specifcity by allostery. Biochem Biophys Res Comm 396, modeling studies. J Med Chem 44, 36-46. 19-23. Pereira, S., Cerqueira, N.M.F.S.A., Fernandes, P.A., and Reichard, P., Baldesten, A., and Rutberg, L. (1961). Ramos, M.J. (2006). Computational studies on class I Formation of deoxycytidine phosphates from cytidine ribonucleotide reductase: Understanding the mechanisms phosphates in extracts from Escherichia coli. J Biol Chem of action and inhibition of a cornerstone enzyme for the 236, 1150. treatment of cancer. Eur Biophys J 35, 125-135. Reichard, P., Eliasson, R., Ingemarson, R., and Thelander, Pereira, S., Fernandes, P.A., and Ramos, M.J. (2004). L. (2000). Cross-talk between the allosteric effector- Mechanism for ribonucleotide reductase inactivation by binding sites in mouse ribonucleotide reductase. J Biol the anticancer drug gemcitabine. J Comput Chem 25, Chem 275, 33021-33026. 1286-1294. 24 Munro and Silva

Richardson, D.R. (2002). Iron chelators as therapeutic Santini, D., Tonini, G., Abbate, A., Di Cosimo, S., Gravante, agents for the treatment of cancer. Crit Rev Oncol Hematol G., Vincenzi, B., Campisi, C., Patti, G., and Di Sciascio, G. 42, 267-281. (2000). Gemcitabine-induced atrial fbrillation: A hitherto Robins, M.J. (1999). Mechanism-based inhibition unreported manifestation of drug toxicity. Ann Oncol 11, of ribonucleotide reductases: New mechanistic 479-481. considerations and promising biological applications. Sato, S., and Wilson, R.J.M. (2005). The plastid of Nucleos Nucleot Nucleic Acids 18, 779-793. Plasmodium spp.: A target for inhibitors. Curr Top Microbiol Rofougaran, R., Vodnala, M., and Hofer, A. (2006). Immunol 295, 251-273. Enzymatically active mammalian ribonucleotide reductase Scherr, M., Steinmann, D., and Eder, M. (2004). RNA exists primarily as an α6β2 octamer. J Biol Chem 281, interference (RNAi) in hematology. Annals of Hematology 27705-27711. 83, 1-8. Roos, D.S., Crawford, M.J., Donald, R.G.K., Fraunholz, Schneider, T.D., and Stephens, R.M. (1990). Sequence M., Harb, O.S., He, C.Y., Kissinger, J.C., Shaw, M.K., logos: a new way to display consensus sequences. and Striepen, B. (2002). Mining the Plasmodium genome Nucleic Acids Res 18, 6097-6100. database to defne organellar function: What does the Shao, J., Zhou, B., Chu, B., and Yen, Y. (2006). Ribonucleotide apicoplast do? Phil Trans Roy Soc Lond B Biol Sci 357, reductase inhibitors and future drug design. Current 35-46. Cancer Drug Targets 6, 409-431. Roos, D.S., Crawford, M.J., Donald, R.G.K., Kissinger, J.C., Singh, B., Kim Sung, L., Matusop, A., Radhakrishnan, Klimczak, L.J., and Striepen, B. (1999). Origin, targeting, A., Shamsul, S.S.G., Cox-Singh, J., Thomas, A., and and function of the apicomplexan plastid. Curr Opin Conway, D.J. (2004). A large focus of naturally acquired Microbiol 2, 426-432. Plasmodium knowlesi infections in human beings. Lancet Roshick, C., Iliffe-Lee, E.R., and McClarty, G. (2000). 363, 1017-1024. Cloning and characterization of ribonucleotide reductase Sjöberg, B.-M. (1997). Ribonucleotide reductases — A from Chlamydia trachomatis. J Biol Chem 275, 38111- group of enzymes with different metallosites and a similar 38119. reaction mechanism. Struct Bond 88, 139-173. Rougemont, M., Van Saanen, M., Sahli, R., Hinrikson, Sjöberg, B.-M. (2010). A Never-Ending Story. Science 329, H.P., Bille, J., and Jaton, K. (2004). Detection of four 1475-1476. Plasmodium species in blood from humans by 18S rRNA Smith, B.D., and Karp, J.E. (2003). Ribonucleotide gene subunit-based and species-specifc real-time PCR reductase: An old target with new potential. Leuk Res 27, assays. J Clin Microbiol 42, 5636-5643. 1075-1076. Rova, U., Adrait, A., Pötsch, S., Gräslund, A., and Sneeden, J.L., and Loeb, L.A. (2004). Mutations in the R2 Thelander, L. (1999). Evidence by mutagenesis that subunit of ribonucleotide reductase that confer resistance Tyr370 of the mouse ribonucleotide reductase R2 protein to hydroxyurea. J Biol Chem 279, 40723-40728. is the connecting link in the intersubunit radical transfer Spielman, D.J. (2009). XVI. Public-private partnerships pathway. J Biol Chem 274, 23746-23751. and pro-poor livestock research: The search for an Rowe, A.K., Rowe, S.Y., Snow, R.W., Korenromp, E.L., East Coast fever vaccine. Enhancing the Effectiveness Schellenberg, J.R.A., Stein, C., Nahlen, B.L., Bryce, J., of Sustainability Partnerships: Summary of a Workshop Black, R.E., and Steketee, R.W. (2006). The burden of National Research Council of the National Academies. malaria mortality among African children in the year 2000. Washinton D.C.: The National Academies Press. 122 p. Int J Epidemiol 35, 691-704. Sridaran, S., Mcclintock, S.K., Syphard, L.M., Herman, Rubin, H., Salem, J.S., Li, L.S., Yang, F.D., Mama, S., K.M., Barnwell, J.W., and Udhayakumar, V. (2010). Anti- Wang, Z.M., Fisher, A., Hamann, C.S., and Cooperman, folate drug resistance in Africa: Meta-analysis of reported B.S. (1993). Cloning, sequence determination, and dihydrofolate reductase (dhfr) and dihydropteroate regulation of the ribonucleotide reductase subunits from synthase (dhps) mutant genotype frequencies in African Plasmodium falciparum: A target for antimalarial therapy. Plasmodium falciparum parasite populations. Malaria Proc Natl Acad Sci Unit States Am 90, 9280-9284. Journal 9, 247. Sachs, J., and Malaney, P. (2002). The economic and social Sriwilaijaroen, N., Boonma, S., Attasart, P., Pothikasikorn, burden of malaria. Nature 415, 680-685. J., Panyim, S., and Noonpakdee, W. (2009). Inhibition Saei, A.A., and Ahmadian, S. (2009). Ribonucleotide of Plasmodium falciparum proliferation in vitro by reductase inhibitors with erythropoietin and iron sulfate double-stranded RNA directed against malaria histone against malaria. Medical hypotheses 72, 611. deacetylase. Biochem Biophys Res Comm 381, 144- Sahu, N.K., Shilakari, G., Nayak, A., and Kohli, D.V. (2007). 147. Antisense technology: A selective tool for gene expression Striepen, B., Pruijssers, A.J.P., Huang, J., Li, C., Gubbels, regulation and gene targeting. Curr Pharmaceut M.-J., Umejiego, N.N., Hedstrom, L., and Kissinger, Biotechnol 8, 291-304. J.C. (2004). Gene transfer in the evolution of parasite Salowe, S., Bollinger, J.M., Ator, M., Stubbe, J., McCraken, nucleotide biosynthesis. Proc Natl Acad Sci Unit States J., Peisach, J., Samano, M.C., and Robins, M.J. (1993). Am 101, 3154-3159. Alternative model for mechanism-based inhibition of Stubbe, J., Nocera, D.G., Yee, C.S., and Chang, M.C.Y. Escherichia coli ribonucleotide reductase by 2'-azido-2'- (2003). Radical initiation in the class I ribonucleotide deoxyuridine 5'-diphosphate. Biochemistry 32, 12749- reductase: Long-range proton-coupled electron transfer? 12760. Chem Rev 103, 2167-2201. Ribonucleotide Reductase as a Target to Control Apicomplexan Diseases 25

Sunil, S., Hossain, M.J., Ramasamy, G., and Malhotra, P. Waller, R.F., and McFadden, G.I. (2005). The apicoplast: A (2008). Transient silencing of Plasmodium falciparum review of the derived plastid of apicomplexan parasites. Tudor Staphylococcal Nuclease suggests an essential Curr Issues Mol Biol 7, 57-79. role for the protein. Biochem Biophys Res Comm 372, Wang, P.J., Chabes, A., Casagrande, R., Tian, X.C., 373-378. Thelander, L., and Huffaker, T.C. (1997). Rnr4p, a novel Szekeres, T., Fritzer-Szekeres, M., and Elford, H.L. (1997). ribonucleotide reductase small-subunit protein. Mol Cell The enzyme ribonucleotide reductase: Target for antitumor Biol 17, 6114-6121. and anti-HIV therapy. Crit Rev Clin Lab Sci 34, 503-528. Weber, J.L. (1987). Analysis of sequences from the Takala, S.L., and Plowe, C.V. (2009). Genetic diversity and extremely A + T-rich genome of Plasmodium falciparum. malaria vaccine design, testing and effcacy: Preventing Gene 52, 103-109. and overcoming 'vaccine resistant malaria'. Parasite Wheeler, L.J., Rajagopal, I., and Mathews, C.K. Immunol 31, 560-573. (2005). Stimulation of mutagenesis by proportional Tanaka, H., Arakawa, H., Yamaguchi, T., Shiraishi, K., deoxyribonucleoside triphosphate accumulation in Fukuda, S., Matsui, K., Takei, Y., and Nakamura, Y. Escherichia coli. DNA Repair 4, 1450-1456. (2000). A ribonucleotide reductase gene involved in a White, N.J. (2004). Antimalarial drug resistance. J Clin p53-dependent cell-cycle checkpoint for DNA damage. Investig 113, 1084-1092. Nature 404, 42-49. Whitnall, M., Howard, J., Ponka, P., and Richardson, D.R. Tedeschi, L., Lande, C., Cecchettini, A., and Citti, L. (2009). (2006). A class of iron chelators with a wide spectrum of Hammerhead ribozymes in therapeutic target discovery potent antitumor activity that overcomes resistance to and validation. Drug Discov Today 14, 776-783. chemotherapeutics. Proc Natl Acad Sci Unit States Am Thelander, L., and Reichard, P. (1979). Reduction of 103, 14901-14906. ribonucleotides. Annu Rev Biochem 48, 133-158. Wiesner, J., Reichenberg, A., Heinrich, S., Schlitzer, M., Ting, L.-M., Shi, W., Lewandowicz, A., Singh, V., Mwakingwe, and Jomaa, H. (2008). The plastid-like organelle of A., Birck, M.R., Ringia, E.A.T., Bench, G., Madrid, D.C., apicomplexan parasites as drug target. Curr Pharmaceut Tyler, P.C., et al. (2005). Targeting a novel Plasmodium Des 14, 855-871. falciparum purine recycling pathway with specifc Winzeler, E.A. (2008). Malaria research in the post-genomic immucillins. J Biol Chem 280, 9547-9554. era. Nature 455, 751-756. Torrents, E., and Sjöberg, B.-M. (2010). Antibacterial activity Xue, X., Zhang, Q., Huang, Y., Feng, L., and Pan, W. of radical scavengers against class Ib ribonucleotide (2008). No miRNA were found in Plasmodium and the reductase from Bacillus anthracis. Biol Chem 391, 229- ones identifed in erythrocytes could not be correlated 234. with infection. Malaria Journal 7, 47. Tracy, S.M., and Sherman, I.W. (1972). Purine uptake and Yanamoto, S., Iwamoto, T., Kawasaki, G., Yoshitomi, I., utilization by the avian malaria parasite Plasmodium Baba, N., and Mizuno, A. (2005). Silencing of the p53R2 lophurae. J Protozool 19, 541-549. gene by RNA interference inhibits growth and enhances Turner, P.C. (2000). Ribozymes. Their design and use in 5-fuorouracil sensitivity of oral cancer cells. Canc Lett cancer. Adv Exp Med Biol 465, 303-318. 223, 67-76. Tuteja, R., Pradhan, A., and Sharma, S. (2008). Plasmodium Yang, F., Curran, S.C., Li, L.S., Avarbock, D., Graf, falciparum signal peptidase is regulated by phosphorylation J.D., Chua, M.M., Lu, G., Salem, J., and Rubin, H. and required for intra-erythrocytic growth. Mol Biochem (1997). Characterization of two genes encoding the Parasitol 157, 137-147. Mycobacterium tuberculosis ribonucleotide reductase Ullu, E., Tschudi, C., and Chakraborty, T. (2004). RNA small subunit. J Bacteriol 179, 6408-6415. interference in protozoan parasites. Cell Microbiol 6, 509- Yang, F., Spanevello, R., Celiker, I., Hirschmann, R., 519. Rubin, H., and Cooperman, B.S. (1990). The carboxyl Uppsten, M., Färnegårdh, M., Domkin, V., and Uhlin, U. terminus heptapeptide of the R2 subunit of mammalian (2006). The frst holocomplex structure of ribonucleotide ribonucleotide reductase inhibits enzyme activity and can reductase gives new insight into its mechanism of action. be used to purify the R1 subunit. FEBS Lett 272, 61-64. J Mol Biol 359, 365-377. Yeh, I., and Altman, R.B. (2006). Drug targets for Plasmodium Vaish, N., Chen, F., Seth, S., Fosnaugh, K., Liu, Y., Adami, falciparum: A post-genomic review/survey. Mini-Rev Med R., Brown, T., Chen, Y., Harvie, P., Johns, R., et al. Chem 6, 177-202. (2010). Improved specifcity of gene silencing by siRNAs Yeh, I., Hanekamp, T., Tsoka, S., Karp, P.D., and Altman, containing unlocked nucleobase analogs. Nucleic Acids R.B. (2004). Computational analysis of Plasmodium Res, 1-10. falciparum metabolism: Organizing genomic information Vedadi, M., Lew, J., Artz, J., Amani, M., Zhao, Y., Dong, A., to facilitate drug discovery. Genome Res 14, 917-924. Wasney, G.A., Gao, M., Hills, T., Brokx, S., et al. (2007). Yen, Y. (2003). Ribonucleotide reductase subunit one as Genome-scale protein expression and structural biology gene therapy target: commentary re: M-Y. Cao et al., of Plasmodium falciparum and related Apicomplexan Adenovirus-mediated ribonucleotide reductase R1 gene organisms. Mol Biochem Parasitol 151, 100-110. therapy of human colon adenocarcinoma. Clin. Cancer Voegtli, W.C., Ge, J., Perlstein, D.L., Stubbe, J., and Res., 9: 4304-4308, 2003. Clin Cancer Res 9, 4304- Rosenzweig, A.C. (2001). Structure of the yeast 4308. ribonucleotide reductase Y2Y4 heterodimer. Proc Natl Acad Sci Unit States Am 98, 10073-10078. Further Reading

Caister Academic Press is a leading academic publisher of advanced texts in microbiology, molecular biology and medical research. Full details of all our publications at caister.com

• MALDI-TOF Mass Spectrometry in Microbiology Edited by: M Kostrzewa, S Schubert (2016) www.caister.com/malditof • Aspergillus and Penicillium in the Post-genomic Era Edited by: RP Vries, IB Gelber, MR Andersen (2016) www.caister.com/aspergillus2

• The Bacteriocins: Current Knowledge and Future Prospects Edited by: RL Dorit, SM Roy, MA Riley (2016) www.caister.com/bacteriocins • Omics in Disease Resistance Edited by: V Bhadauria (2016) www.caister.com/opdr

• Acidophiles: Life in Extremely Acidic Environments Edited by: R Quatrini, DB Johnson (2016) www.caister.com/acidophiles

• Climate Change and Microbial Ecology: Current Research and Future Trends Edited by: J Marxsen (2016) www.caister.com/climate

• Biofilms in Bioremediation: Current Research and Emerging Technologies • Flow Cytometry in Microbiology: Technology and Applications Edited by: G Lear (2016) www.caister.com/biorem Edited by: MG Wilkinson (2015) www.caister.com/flow • Microalgae: Current Research and Applications • Probiotics and Prebiotics: Current Research and Future Trends Edited by: MN Tsaloglou (2016) www.caister.com/microalgae Edited by: K Venema, AP Carmo (2015) www.caister.com/probiotics • Gas Plasma Sterilization in Microbiology: Theory, Applications, Pitfalls and New Perspectives • Epigenetics: Current Research and Emerging Trends

Edited by: H Shintani, A Sakudo (2016) Edited by: BP Chadwick (2015) www.caister.com/gasplasma www.caister.com/epigenetics2015

• Virus Evolution: Current Research and Future Directions • Corynebacterium glutamicum: From Systems Biology to Biotechnological Applications Edited by: SC Weaver, M Denison, M Roossinck, et al. (2016) www.caister.com/virusevol Edited by: A Burkovski (2015) www.caister.com/cory2 • Arboviruses: Molecular Biology, Evolution and Control Edited by: N Vasilakis, DJ Gubler (2016) • Advanced Vaccine Research Methods for the Decade of www.caister.com/arbo Vaccines Edited by: F Bagnoli, R Rappuoli (2015) • Shigella: Molecular and Cellular Biology www.caister.com/vaccines Edited by: WD Picking, WL Picking (2016) www.caister.com/shigella • Antifungals: From Genomics to Resistance and the Development of Novel Agents

• Aquatic Biofilms: Ecology, Water Quality and Wastewater Edited by: AT Coste, P Vandeputte (2015) Treatment www.caister.com/antifungals Edited by: AM Romaní, H Guasch, MD Balaguer (2016) www.caister.com/aquaticbiofilms • Bacteria-Plant Interactions: Advanced Research and Future Trends Edited by: J Murillo, BA Vinatzer, RW Jackson, et al. (2015) • Alphaviruses: Current Biology www.caister.com/bacteria-plant Edited by: S Mahalingam, L Herrero, B Herring (2016) www.caister.com/alpha • Aeromonas Edited by: J Graf (2015) • Thermophilic Microorganisms www.caister.com/aeromonas Edited by: F Li (2015) www.caister.com/thermophile • Antibiotics: Current Innovations and Future Trends Edited by: S Sánchez, AL Demain (2015) www.caister.com/antibiotics

• Leishmania: Current Biology and Control Edited by: S Adak, R Datta (2015) www.caister.com/leish2

• Acanthamoeba: Biology and Pathogenesis (2nd edition) Author: NA Khan (2015) www.caister.com/acanthamoeba2

• Microarrays: Current Technology, Innovations and Applications Edited by: Z He (2014) www.caister.com/microarrays2

• Metagenomics of the Microbial Nitrogen Cycle: Theory, Methods and Applications Edited by: D Marco (2014) www.caister.com/n2

Order from caister.com/order